ANTI-TCR ANTIBODY MOLECULES AND USES THEREOF

The disclosure provides antibody molecules that bind to TCR Vβ regions and multispecific molecules comprising said antibody molecules. Additionally, disclosed are nucleic acids encoding the same, methods of producing the aforesaid molecules, pharmaceutical compositions comprising aforesaid molecules, and methods of treating a cancer using the aforesaid molecules.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. application Ser. No. 17/855,332 filed Jun. 30, 2022, which is a continuation of International Application No. PCT/US2020/067543, filed on Dec. 30, 2020, which claims the benefit of U.S. Provisional Application 62/957,024 filed on Jan. 3, 2020, and U.S. Provisional Application 63/070,596 filed on Aug. 26, 2020, the entire contents of each of which are hereby incorporated by reference.

SEQUENCE LISTING

This application contains a Sequence Listing which has been submitted electronically in XML format. The Sequence Listing XML is incorporated herein by reference. Said XML file, created on Apr. 13, 2023, is named 53676-736_302_SL.xml and is 1,498,604 bytes in size.

BACKGROUND

Current molecules designed to redirect T cells to promote tumor cell lysis for cancer immunotherapy typically target the CD3 epsilon (CD3e) subunit of the T cell receptor (TCR). However, there are limitations to this approach. Previous studies have shown that, e.g., low doses of anti-CD3e monoclonal antibody (mAb) can cause T cell dysfunction and exert immunosuppressive effects. In addition, anti-CD3e mAbs bind to all T cells and thus activate a large number of T cells. Such non-physiological massive activation of T cells by these anti-CD3e mAbs can result in the production of proinflammatory cytokines such as IFN-gamma, IL-1-beta, IL-6, IL-10 and TNF-alpha, causing a “cytokine storm” known as the cytokine release syndrome (CRS), which is also associated with neurotoxicity (NT). Thus, it might be advantageous to develop antibodies that avoid or reduce CRS and/or NT.

SUMMARY OF THE INVENTION

In an aspect, provided herein is, inter alia, a composition comprising a molecule that comprises an antigen binding domain that binds to a T cell receptor beta variable (TCRβV) region, wherein the antigen binding domain comprises:

    • (A) a heavy chain variable region (VH) comprising:
    • (a) a heavy chain complementarity determining region 3 (HC CDR3) comprising the sequence SYYSYDVLDY (SEQ ID NO: 47); and
    • (i) a heavy chain complementarity determining region 1 (HC CDR1) comprising the sequence TYYIH (SEQ ID NO: 45), and a heavy chain complementarity determining region 2 (HC CDR2) comprising the sequence WFFPGSGNIKYNEKFKG (SEQ ID NO: 46);
    • (ii) a HC CDR1 comprising the sequence GHDFRLTYIH (amino acids 26-35 of SEQ ID NO: 1346), and a HC CDR2 comprising the sequence RVSAGSGNVKYNEKFKG (amino acids 50-66 of SEQ ID NO: 1346);
    • (iii) a HC CDR1 comprising the sequence GHDFRLTYIH (amino acids 26-35 of SEQ ID NO: 1356), and a HC CDR2 comprising the sequence RVYAGSGNTKYNEKFKG (amino acids 50-66 of SEQ ID NO: 1356); or
    • (iv) a HC CDR1 comprising the sequence GHDFKLTYIH (amino acids 26-35 of SEQ ID NO: 1370), and a HC CDR2 comprising the sequence RVSAGSGNVNYAQKFQG (amino acids 50-66 of SEQ ID NO: 1370); or
    • (b) a HC CDR3 comprising the sequence of SLNWDYGLDY (SEQ ID NO: 1104); and
    • (i) a HC CDR1 comprising the sequence SAYMH (SEQ ID NO: 1102), and a HC CDR2 comprising the sequence RIDPATGKTKYAPKFQA (SEQ ID NO: 1103); or
    • (ii) a HC CDR1 comprising the sequence DTYMY (SEQ ID NO: 1120); and a HC CDR2 comprising the sequence RIDPANGNTKYDPKFQD (SEQ ID NO: 1121); and
    • (B) a light chain variable region (VL) comprising:
    • (a) a light chain complementarity determining region 3 (LC CDR3) comprising the sequence QQFKSYPLT (SEQ ID NO: 53); and
    • (i) a light chain complementarity determining region 1 (LC CDR1) comprising the sequence KASQNVGINVV (SEQ ID NO: 51), and a light chain complementarity determining region 2 (LC CDR2) comprising the sequence SSSHRYS (SEQ ID NO: 52);
    • (ii) a LC CDR1 comprising the sequence KASQNVADRVV (amino acids 24-34 of SEQ ID NO: 1349), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349);
    • (iii) a LC CDR1 comprising the sequence KASQNVEDRVV (amino acids 24-34 of SEQ ID NO: 1357), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349); or
    • (iv) a LC CDR1 comprising the sequence RASQNVDNRLG (amino acids 24-34 of SEQ ID NO: 1365), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349);
    • (b) a LC CDR3 comprising the sequence QQFTSSPFT (SEQ ID NO: 65), a LC CDR1 comprising the sequence RASSSVNYIY (SEQ ID NO: 63), and the LC CDR2 comprising the sequence YTSNLAP (SEQ ID NO: 64);
    • (c) a LC CDR3 comprising the sequence QQSIEDPWT (SEQ ID NO: 1109), a LC CDR1 comprising the sequence RASKSVSILGTHLIH (SEQ ID NO: 1107), and the LC CDR2 comprising the sequence AASNLES (SEQ ID NO: 1108);
    • (d) a LC CDR3 comprising the sequence QQSNEDPYT (SEQ ID NO:1128), a LC CDR1 comprising the sequence RASESVDSYGNSFMH (SEQ ID NO: 1126), and the LC CDR2 comprising the sequence RASNLES (SEQ ID NO:1127);

In some embodiments, the VH comprises:

    • (i) the HC CDR3 comprising the sequence SYYSYDVLDY (SEQ ID NO: 47);
    • (ii) the HC CDR1 comprising the sequence TYYIH (SEQ ID NO: 45); and
    • (iii) the HC CDR2 comprising the sequence WFFPGSGNIKYNEKFKG (SEQ ID NO: 46).

In some embodiments, the VH comprises:

    • (i) the HC CDR3 comprising the sequence SYYSYDVLDY (SEQ ID NO: 47);
    • (ii) the HC CDR1 comprising the sequence GHDFRLTYIH (amino acids 26-35 of SEQ ID NO: 1346); and
    • (iii) the HC CDR2 comprising the sequence RVSAGSGNVKYNEKFKG (amino acids 50-66 of SEQ ID NO: 1346).

In some embodiments, the VH comprises:

    • (i) the HC CDR3 comprising the sequence SYYSYDVLDY (SEQ ID NO: 47);
    • (ii) the HC CDR1 comprising the sequence GHDFRLTYIH (amino acids 26-35 of SEQ ID NO: 1356); and
    • (iii) the HC CDR2 comprising the sequence RVYAGSGNTKYNEKFKG (amino acids 50-66 of SEQ ID NO: 1356).

In some embodiments, the VH comprises:

    • (i) the HC CDR3 comprising the sequence SYYSYDVLDY (SEQ ID NO: 47);
    • (ii) the HC CDR1 comprising the sequence GHDFKLTYIH (amino acids 26-35 of SEQ ID NO: 1370); and
    • (iii) the HC CDR2 comprising the sequence RVSAGSGNVNYAQKFQG (amino acids 50-66 of SEQ ID NO: 1370).

In some embodiments, the VH comprises:

    • (i) the HC CDR3 comprising the sequence SLNWDYGLDY (SEQ ID NO: 1104);
    • (ii) the HC CDR1 comprising the sequence SAYMH (SEQ ID NO: 1102); and
    • (iii) the HC CDR2 comprising the sequence RIDPATGKTKYAPKFQA (SEQ ID NO: 1103).

In some embodiments, the VH comprises:

    • (i) the HC CDR3 comprising the sequence SLNWDYGLDY (SEQ ID NO: 1104);
    • (ii) the HC CDR1 comprising the sequence DTYMY (SEQ ID NO: 1120); and
    • (iii) the HC CDR2 comprising the sequence RIDPANGNTKYDPKFQD (SEQ ID NO: 1121).

In some embodiments, the VL comprises:

    • (A) (i) the LC CDR3 comprising the sequence QQSIEDPWT (SEQ ID NO:1109);
    • (ii) the LC CDR1 comprising the sequence RASKSVSILGTHLIH (SEQ ID NO:1107); and
    • (iii) the LC CDR2 comprising the sequence AASNLES (SEQ ID NO: 1108);
    • (B) (i) the LC CDR3 comprising the sequence QQSNEDPYT (SEQ ID NO:1128);
    • (ii) the LC CDR1 comprising the sequence RASESVDSYGNSFMH (SEQ ID NO:1126); and
    • (iii) the LC CDR2 comprising the sequence RASNLES (SEQ ID NO:1127);
    • (C) (i) the LC CDR3 comprising the sequence QQFKSYPLT (SEQ ID NO: 53);
    • (ii) the LC CDR1 comprising the sequence KASQNVGINVV (SEQ ID NO: 51); and
    • (iii) the LC CDR2 comprising the sequence SSSHRYS (SEQ ID NO: 52);
    • (D) (i) the LC CDR3 comprising the sequence QQFTSSPFT (SEQ ID NO: 65);
    • (ii) the LC CDR1 comprising the sequence RASSSVNYIY (SEQ ID NO: 63); and
    • (iii) the LC CDR2 comprising the sequence YTSNLAP (SEQ ID NO: 64);
    • (E) (i) the LC CDR3 comprising the sequence QQFKSYPLT (SEQ ID NO: 53);
    • (ii) the LC CDR1 comprising the sequence KASQNVADRVV (amino acids 24-34 of SEQ ID NO: 1349); and
    • (iii) the LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349);
    • (F) (i) the LC CDR3 comprising the sequence QQFKSYPLT (SEQ ID NO: 53);
    • (ii) the LC CDR1 comprising the sequence KASQNVEDRVV (amino acids 24-34 of SEQ ID NO: 1357); and
    • (iii) the LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349); or
    • (G) (i) the LC CDR3 comprising the sequence QQFKSYPLT (SEQ ID NO: 53);
    • (ii) the LC CDR1 comprising the sequence RASQNVDNRLG (amino acids 24-34 of SEQ ID NO: 1365); and
    • (iii) the LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349).

In some embodiments, the VL comprises:

    • (i) the LC CDR3 comprising the sequence QQFKSYPLT (SEQ ID NO: 53);
    • (ii) the LC CDR1 comprising the sequence KASQNVGINVV (SEQ ID NO: 51); and
    • (iii) the LC CDR2 comprising the sequence SSSHRYS (SEQ ID NO: 52).

In some embodiments, the VL comprises:

    • (i) the LC CDR3 comprising the sequence QQFKSYPLT (SEQ ID NO: 53);
    • (ii) the LC CDR1 comprising the sequence KASQNVADRVV (amino acids 24-34 of SEQ ID NO: 1349); and
    • (iii) the LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349).

In some embodiments, the VL comprises:

    • (i) the LC CDR3 comprising the sequence QQFKSYPLT (SEQ ID NO: 53);
    • (ii) the LC CDR1 comprising the sequence KASQNVEDRVV (amino acids 24-34 of SEQ ID NO: 1357); and
    • (iii) the LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349).

In some embodiments, the VL comprises:

    • (i) the LC CDR3 comprising the sequence QQFKSYPLT (SEQ ID NO: 53);
    • (ii) the LC CDR1 comprising the sequence RASQNVDNRLG (amino acids 24-34 of SEQ ID NO: 1365); and
    • (iii) the LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349).

In some embodiments, the VL comprises:

    • (i) the LC CDR3 comprising the sequence QQSIEDPWT (SEQ ID NO:1109);
    • (ii) a LC CDR1 comprising the sequence RASKSVSILGTHLIH (SEQ ID NO:1107); and
    • (iii) a LC CDR2 comprising the sequence AASNLES (SEQ ID NO: 1108).

In some embodiments, the antigen binding domain comprises:

    • (i) a VH comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1100, and a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1101;
    • (ii) a VH comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1346, and a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1349;
    • (iii) a VH comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1356, and a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1357;
    • (iv) a VH comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1370, and a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1365; or
    • (v) a VH comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1112, SEQ ID NO: 1113, SEQ ID NO: 1114, SEQ ID NO: 1115, or SEQ ID NO: 1116, and a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1111, SEQ ID NO: 1117, SEQ ID NO: 1118, or SEQ ID NO: 1119.

In some embodiments, the antigen binding domain comprises:

    • (i) a VH comprising the sequence of SEQ ID NO: 1100, and a VL comprising the sequence of SEQ ID NO: 1101;
    • (ii) a VH comprising the sequence of SEQ ID NO: 1346, and a VL comprising the sequence of SEQ ID NO: 1349;
    • (iii) a VH comprising the sequence of SEQ ID NO: 1356, and a VL comprising the sequence of SEQ ID NO: 1357;
    • (iv) a VH comprising the sequence of SEQ ID NO: 1370, and a VL comprising the sequence of SEQ ID NO: 1365; or
    • (v) a VH comprising the sequence of SEQ ID NO: 1112, SEQ ID NO: 1113, SEQ ID NO: 1114, SEQ ID NO: 1115, or SEQ ID NO: 1116, and a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1111, SEQ ID NO: 1117, SEQ ID NO: 1118, or SEQ ID NO: 1119.

In some embodiments, the antigen binding domain is a Fab or a single chain Fv (scFv).

In some embodiments, the molecule comprises at least two non-contiguous polypeptide chains, wherein a first polypeptide chain of the at least two non-contiguous polypeptide chains comprises a first Fc region, and a second polypeptide chain of the at least two non-contiguous polypeptide chains comprises a second Fc region; and wherein the first Fc region and the second Fc region comprise an Fc interface with a knob-in-a hole.

In some embodiments, the molecule comprises the first polypeptide chain, the second polypeptide chain, and a third polypeptide chain, wherein the first polypeptide chain, the second polypeptide chain, and the third polypeptide chain are non-contiguous, and wherein:

    • (i) the first polypeptide chain comprises a VH comprising the sequence of SEQ ID NO: 1346, wherein the VH is linked to the first Fc region,
    • (ii) the second polypeptide chain comprises a cytokine molecule,
    • wherein the cytokine molecule is linked to the second Fc region, and
    • wherein the cytokine molecule comprises: (a) IL-2 or functional variant thereof comprising the sequence of SEQ ID NO: 2270, or (b) IL-15 or functional variant thereof comprising the sequence of SEQ ID NO: 2170, wherein the IL-15 or functional variant thereof is covalently linked to an IL15Ralpha dimerizing domain, and
    • (iii) the third polypeptide chain comprises a VL comprising the sequence of SEQ ID NO: 1349, wherein the VL is linked to a light chain constant region comprising the sequence of SEQ ID NO: 39.

In some embodiments, the molecule comprises the first polypeptide chain, the second polypeptide chain, and a third polypeptide chain, wherein the first polypeptide chain, the second polypeptide chain, and the third polypeptide chain are non-contiguous, and wherein:

    • (i) the first polypeptide chain comprises a VH comprising the sequence of SEQ ID NO: 1113, wherein the VH is linked to the first Fc region,
    • (ii) the second polypeptide chain comprises a cytokine molecule,
    • wherein the cytokine molecule is linked to the second Fc region, and
    • wherein the cytokine molecule comprises: (a) IL-2 or functional variant thereof comprising the sequence of SEQ ID NO: 2270, or (b) IL-15 or functional variant thereof comprising the sequence of SEQ ID NO: 2170, wherein the IL-15 or functional variant thereof is covalently linked to an IL15Ralpha dimerizing domain, and
    • (iii) the third polypeptide chain comprises a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1111, wherein the VL is linked to a light chain constant region comprising the sequence of SEQ ID NO: 39.

In some embodiments, the molecule comprises the first polypeptide chain, the second polypeptide chain, and a third polypeptide chain, wherein the first polypeptide chain, the second polypeptide chain, and the third polypeptide chain are non-contiguous, and wherein:

    • (i) the first polypeptide chain comprises a VH comprising the sequence of SEQ ID NO: 1100, wherein the VH is linked to the first Fc region,
    • (ii) the second polypeptide chain comprises a cytokine molecule,
    • wherein the cytokine molecule is linked to the second Fc region, and
    • wherein the cytokine molecule comprises: (a) IL-2 or functional variant thereof comprising the sequence of SEQ ID NO: 2270, or (b) IL-15 or functional variant thereof comprising the sequence of SEQ ID NO: 2170, wherein the IL-15 or functional variant thereof is covalently linked to an IL15Ralpha dimerizing domain, and
    • (iii) the third polypeptide chain comprises a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1101, wherein the VL is linked to a light chain constant region comprising the sequence of SEQ ID NO: 39.

In some embodiments, the molecule comprises the first polypeptide chain, the second polypeptide chain, and a third polypeptide chain, wherein the first polypeptide chain, the second polypeptide chain, and the third polypeptide chain are non-contiguous, and wherein:

    • (i) the first polypeptide chain comprises a VH comprising the sequence of SEQ ID NO: 1356, wherein the VH is linked to the first Fc region,
    • (ii) the second polypeptide chain comprises a cytokine molecule,
    • wherein the cytokine molecule is linked to the second Fc region, and
    • wherein the cytokine molecule comprises: (a) IL-2 or functional variant thereof comprising the sequence of SEQ ID NO: 2270, or (b) IL-15 or functional variant thereof comprising the sequence of SEQ ID NO: 2170, wherein the IL-15 or functional variant thereof is covalently linked to an IL15Ralpha dimerizing domain, and
    • (iii) the third polypeptide chain comprises a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1357, wherein the VL is linked to a light chain constant region comprising the sequence of SEQ ID NO: 39.

In some embodiments, the molecule comprises the first polypeptide chain, the second polypeptide chain, and a third polypeptide chain, wherein the first polypeptide chain, the second polypeptide chain, and the third polypeptide chain are non-contiguous, and wherein:

    • (i) the first polypeptide chain comprises a VH comprising the sequence of SEQ ID NO: 1370, wherein the VH is linked to the first Fc region,
    • (ii) the second polypeptide chain comprises a cytokine molecule,
    • wherein the cytokine molecule is linked to the second Fc region,
    • wherein the cytokine molecule comprises: (a) IL-2 or functional variant thereof comprising the sequence of SEQ ID NO: 2270, or (b) IL-15 or functional variant thereof comprising the sequence of SEQ ID NO: 2170, wherein the IL-15 or functional variant thereof is covalently linked to an IL15Ralpha dimerizing domain, and
    • (iii) the third polypeptide chain comprises a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1365, wherein the VL is linked to a light chain constant region comprising the sequence of SEQ ID NO: 39.

In some embodiments, (1) the first Fc region and the second Fc region each comprises an Asn297Ala mutation;

    • (2) (i) the first Fc region and the second Fc region each comprises a sequence having at least 98% sequence identity to SEQ ID NO: 41 or SEQ ID NO: 42, or
    • (ii) (a) the first Fc region comprising the sequence of amino acids 1-328 of SEQ ID NO: 42, with the proviso that Tyr at the position 349, Thr at the position 366, Leu at the position 368, and Tyr at the position 407 according to EU Numbering are replaced with Cys, Ser, Ala, and Val, respectively; and
    • (b) the second polypeptide chain comprises the cytokine molecule linked to the second Fc region via the sequence of SEQ ID NO: 3308, wherein the second Fc region comprises the sequence of amino acids 104-328 of SEQ ID NO: 42, with the proviso that Ser at the position 354 and Thr at the position 366 according to EU Numbering are replaced with Cys and Trp, respectively; or
    • (3) any combination thereof.

In some embodiments, (1) the first Fc region and the second Fc region each comprises an Asn297Ala mutation;

    • (2) (i) the first Fc region and the second Fc region each comprises a sequence having at least 98% sequence identity to SEQ ID NO: 41 or SEQ ID NO: 42, or
    • (ii) (a) the first Fc region comprising the sequence of amino acids 1-328 of SEQ ID NO: 42, with the proviso that Tyr at the position 349, Thr at the position 366, Leu at the position 368, and Tyr at the position 407 according to EU Numbering are replaced with Cys, Ser, Ala, and Val, respectively; and
    • (b) the second polypeptide chain comprises the cytokine molecule linked to the second Fc region via the sequence of SEQ ID NO: 3308, wherein the second Fc region comprises the sequence of amino acids 104-328 of SEQ ID NO: 42, with the proviso that Ser at the position 354 and Thr at the position 366 according to EU Numbering are replaced with Cys and Trp, respectively; or (3) any combination thereof.

In some embodiments, (1) the first Fc region and the second Fc region each comprises an Asn297Ala mutation;

    • (2) (i) the first Fc region and the second Fc region each comprises a sequence having at least 98% sequence identity to SEQ ID NO: 41 or SEQ ID NO: 42, or
    • (ii) (a) the first Fc region comprising the sequence of amino acids 1-328 of SEQ ID NO: 42, with the proviso that Tyr at the position 349, Thr at the position 366, Leu at the position 368, and Tyr at the position 407 according to EU Numbering are replaced with Cys, Ser, Ala, and Val, respectively; and
    • (b) the second polypeptide chain comprises the cytokine molecule linked to the second Fc region via the sequence of SEQ ID NO: 3308, wherein the second Fc region comprises the sequence of amino acids 104-328 of SEQ ID NO: 42, with the proviso that Ser at the position 354 and Thr at the position 366 according to EU Numbering are replaced with Cys and Trp, respectively; or (3) any combination thereof.

In some embodiments, (1) the first Fc region and the second Fc region each comprises an Asn297Ala mutation;

    • (2) (i) the first Fc region and the second Fc region each comprises a sequence having at least 98% sequence identity to SEQ ID NO: 41 or SEQ ID NO: 42, or
    • (ii) (a) the first Fc region comprising the sequence of amino acids 1-328 of SEQ ID NO: 42, with the proviso that Tyr at the position 349, Thr at the position 366, Leu at the position 368, and Tyr at the position 407 according to EU Numbering are replaced with Cys, Ser, Ala, and Val, respectively; and
    • (b) the second polypeptide chain comprises the cytokine molecule linked to the second Fc region via the sequence of SEQ ID NO: 3308, wherein the second Fc region comprises the sequence of amino acids 104-328 of SEQ ID NO: 42, with the proviso that Ser at the position 354 and Thr at the position 366 according to EU Numbering are replaced with Cys and Trp, respectively; or
    • (3) any combination thereof.

In some embodiments, (1) the first Fc region and the second Fc region each comprises an Asn297Ala mutation;

    • (2) (i) the first Fc region and the second Fc region each comprises a sequence having at least 98% sequence identity to SEQ ID NO: 41 or SEQ ID NO: 42, or
    • (ii) (a) the first Fc region comprising the sequence of amino acids 1-328 of SEQ ID NO: 42, with the proviso that Tyr at the position 349, Thr at the position 366, Leu at the position 368, and Tyr at the position 407 according to EU Numbering are replaced with Cys, Ser, Ala, and Val, respectively; and
    • (b) the second polypeptide chain comprises the cytokine molecule linked to the second Fc region via the sequence of SEQ ID NO: 3308, wherein the second Fc region comprises the sequence of amino acids 104-328 of SEQ ID NO: 42, with the proviso that Ser at the position 354 and Thr at the position 366 according to EU Numbering are replaced with Cys and Trp, respectively; or
    • (3) any combination thereof.

In some embodiments, the second polypeptide chain comprises the antigen binding domain comprising the sequence of SEQ ID NO: 1331, and a cytokine molecule, wherein the cytokine molecule comprises IL-2 or functional variant thereof comprising the sequence of SEQ ID NO: 2270, and wherein the antigen binding domain, the cytokine molecule, and the second Fc region are linked.

In another aspect, provided herein is a method of treating cancer in a human subject in need thereof comprising administering to the human subject a therapeutically effective amount of a molecule comprising an anti-gen binding domain that binds to a T cell receptor beta variable (TCRβV) region, wherein the antigen binding domain comprises:

    • (A) a heavy chain variable region (VH) comprising:
    • (a) a heavy chain complementarity determining region 3 (HC CDR3) comprising the sequence SYYSYDVLDY (SEQ ID NO: 47); and
    • (i) a heavy chain complementarity determining region 1 (HC CDR1) comprising the sequence TYYIH (SEQ ID NO: 45), and a heavy chain complementarity determining region 2 (HC CDR2) comprising the sequence WFFPGSGNIKYNEKFKG (SEQ ID NO: 46);
    • (ii) a HC CDR1 comprising the sequence GHDFRLTYIH (amino acids 26-35 of SEQ ID NO: 1346), and a HC CDR2 comprising the sequence RVSAGSGNVKYNEKFKG (amino acids 50-66 of SEQ ID NO: 1346);
    • (iii) a HC CDR1 comprising the sequence GHDFRLTYIH (amino acids 26-35 of SEQ ID NO: 1356), and a HC CDR2 comprising the sequence RVYAGSGNTKYNEKFKG (amino acids 50-66 of SEQ ID NO: 1356); or
    • (iv) a HC CDR1 comprising the sequence GHDFKLTYIH (amino acids 26-35 of SEQ ID NO: 1370), and a HC CDR2 comprising the sequence RVSAGSGNVNYAQKFQG (amino acids 50-66 of SEQ ID NO: 1370); or
    • (b) a HC CDR3 comprising the sequence of SLNWDYGLDY (SEQ ID NO: 1104); and
    • (i) a HC CDR1 comprising the sequence SAYMH (SEQ ID NO: 1102), and a HC CDR2 comprising the sequence RIDPATGKTKYAPKFQA (SEQ ID NO: 1103); or
    • (ii) a HC CDR1 comprising the sequence DTYMY (SEQ ID NO: 1120); and a HC CDR2 comprising the sequence RIDPANGNTKYDPKFQD (SEQ ID NO: 1121); and
    • (B) a light chain variable region (VL) comprising:
    • (a) a light chain complementarity determining region 3 (LC CDR3) comprising the sequence QQFKSYPLT (SEQ ID NO: 53); and
    • (i) a light chain complementarity determining region 1 (LC CDR1) comprising the sequence KASQNVGINVV (SEQ ID NO: 51), and a light chain complementarity determining region 2 (LC CDR2) comprising the sequence SSSHRYS (SEQ ID NO: 52);
    • (ii) a LC CDR1 comprising the sequence KASQNVADRVV (amino acids 24-34 of SEQ ID NO: 1349), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349);
    • (iii) a LC CDR1 comprising the sequence KASQNVEDRVV (amino acids 24-34 of SEQ ID NO: 1357), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349); or
    • (iv) a LC CDR1 comprising the sequence RASQNVDNRLG (amino acids 24-34 of SEQ ID NO: 1365), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349);
    • (b) a LC CDR3 comprising the sequence QQFTSSPFT (SEQ ID NO: 65), a LC CDR1 comprising the sequence RASSSVNYIY (SEQ ID NO: 63), and the LC CDR2 comprising the sequence YTSNLAP (SEQ ID NO: 64);
    • (c) a LC CDR3 comprising the sequence QQSIEDPWT (SEQ ID NO: 1109), a LC CDR1 comprising the sequence RASKSVSILGTHLIH (SEQ ID NO: 1107), and the LC CDR2 comprising the sequence AASNLES (SEQ ID NO: 1108);
    • (d) a LC CDR3 comprising the sequence QQSNEDPYT (SEQ ID NO:1128), a LC CDR1 comprising the sequence RASESVDSYGNSFMH (SEQ ID NO: 1126), and the LC CDR2 comprising the sequence RASNLES (SEQ ID NO:1127), thereby treating the cancer in the human subject.

In another aspect, provided herein is a method of expanding an immune cell population comprising contacting the immune cell population with a composition comprising a molecule comprising an antigen binding domain that binds to a T cell receptor beta variable (TCRβV) region, wherein the antigen binding domain comprises:

    • (A) a heavy chain variable region (VH) comprising:
    • (a) a heavy chain complementarity determining region 3 (HC CDR3) comprising the sequence SYYSYDVLDY (SEQ ID NO: 47); and
    • (i) a heavy chain complementarity determining region 1 (HC CDR1) comprising the sequence TYYIH (SEQ ID NO: 45), and a heavy chain complementarity determining region 2 (HC CDR2) comprising the sequence WFFPGSGNIKYNEKFKG (SEQ ID NO: 46);
    • (ii) a HC CDR1 comprising the sequence GHDFRLTYIH (amino acids 26-35 of SEQ ID NO: 1346), and a HC CDR2 comprising the sequence RVSAGSGNVKYNEKFKG (amino acids 50-66 of SEQ ID NO: 1346);
    • (iii) a HC CDR1 comprising the sequence GHDFRLTYIH (amino acids 26-35 of SEQ ID NO: 1356), and a HC CDR2 comprising the sequence RVYAGSGNTKYNEKFKG (amino acids 50-66 of SEQ ID NO: 1356); or
    • (iv) a HC CDR1 comprising the sequence GHDFKLTYIH (amino acids 26-35 of SEQ ID NO: 1370), and a HC CDR2 comprising the sequence RVSAGSGNVNYAQKFQG (amino acids 50-66 of SEQ ID NO: 1370); or
    • (b) a HC CDR3 comprising the sequence of SLNWDYGLDY (SEQ ID NO: 1104); and
    • (i) a HC CDR1 comprising the sequence SAYMH (SEQ ID NO: 1102), and a HC CDR2 comprising the sequence RIDPATGKTKYAPKFQA (SEQ ID NO: 1103); or
    • (ii) a HC CDR1 comprising the sequence DTYMY (SEQ ID NO: 1120); and a HC CDR2 comprising the sequence RIDPANGNTKYDPKFQD (SEQ ID NO: 1121); and
    • (B) a light chain variable region (VL) comprising:
    • (a) a light chain complementarity determining region 3 (LC CDR3) comprising the sequence QQFKSYPLT (SEQ ID NO: 53); and
    • (i) a light chain complementarity determining region 1 (LC CDR1) comprising the sequence KASQNVGINVV (SEQ ID NO: 51), and a light chain complementarity determining region 2 (LC CDR2) comprising the sequence SSSHRYS (SEQ ID NO: 52);
    • (ii) a LC CDR1 comprising the sequence KASQNVADRVV (amino acids 24-34 of SEQ ID NO: 1349), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349);
    • (iii) a LC CDR1 comprising the sequence KASQNVEDRVV (amino acids 24-34 of SEQ ID NO: 1357), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349); or
    • (iv) a LC CDR1 comprising the sequence RASQNVDNRLG (amino acids 24-34 of SEQ ID NO: 1365), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349);
    • (b) a LC CDR3 comprising the sequence QQFTSSPFT (SEQ ID NO: 65), a LC CDR1 comprising the sequence RASSSVNYIY (SEQ ID NO: 63), and the LC CDR2 comprising the sequence YTSNLAP (SEQ ID NO: 64);
    • (c) a LC CDR3 comprising the sequence QQSIEDPWT (SEQ ID NO: 1109), a LC CDR1 comprising the sequence RASKSVSILGTHLIH (SEQ ID NO: 1107), and the LC CDR2 comprising the sequence AASNLES (SEQ ID NO: 1108);
    • (d) a LC CDR3 comprising the sequence QQSNEDPYT (SEQ ID NO:1128), a LC CDR1 comprising the sequence RASESVDSYGNSFMH (SEQ ID NO: 1126), and the LC CDR2 comprising the sequence RASNLES (SEQ ID NO:1127),
    • thereby expanding the immune cell population, and
    • wherein the expansion occurs in vivo or ex vivo.

BRIEF DESCRIPTION OF THE DRAWINGS

The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.

FIGS. 1A-1B shows the alignment of the Antibody A source mouse VH and VL framework 1, CDR 1, framework 2, CDR 2, framework 3, CDR3, and framework 4 regions with their respective humanized sequences. Kabat CDRs are shown in bold, Chothia CDRs are shown in italics, and combined CDRs are shown in boxes. The framework positions that were back mutated are double underlined. FIG. 1A shows VH sequences for murine Antibody A (SEQ ID NO: 1) and humanized Antibody A-H (SEQ ID NO: 9). FIG. 1B shows VL sequences for murine Antibody A (SEQ ID NO: 2) and humanized Antibody A-H (SEQ ID NO: 10 and SEQ ID NO: 11).

FIGS. 2A-2C shows the alignment of the Antibody B source mouse VH and VL framework 1, CDR 1, framework 2, CDR 2, framework 3, CDR3, and framework 4 regions with their respective humanized sequences. Kabat CDRs are shown in bold, Chothia CDRs are shown in italics, and combined CDRs are shown in boxes. The framework positions that were back mutated are double underlined. FIG. 2A shows the VH sequence for murine Antibody B (SEQ ID NO: 15) and humanized VH sequences B-H.1A to B-H.1C (SEQ ID NOs: 23-25). FIG. 2B shows the VL sequence for murine Antibody B (SEQ ID NO: 16) and humanized VL sequences B-H.1D to B-H.1H (SEQ ID NOs: 26-30). FIG. 2C shows the VL sequence for murine Antibody B (SEQ ID NO: 16) and humanized VL sequences B-H.1D to B-H.1H (SEQ ID NOs: 26-30) continued from FIG. 2B.

FIG. 3 depicts the phylogenetic tree of TCRBV gene family and subfamilies with corresponding antibodies mapped. Subfamily identities are as follows: Subfamily A: TCRβ V6; Subfamily B: TCRβ V10; Subfamily C: TCRβ V12; Subfamily D: TCRβ V5; Subfamily E: TCRβ V7; Subfamily F: TCRβ V11; Subfamily G: TCRβ V14; Subfamily H: TCRβ V16; Subfamily I:TCRβ V18; Subfamily J:TCRβ V9; Subfamily K: TCRβ V13; Subfamily L: TCRβ V4; Subfamily M:TCRβ V3; Subfamily N:TCRβ V2; Subfamily O:TCRβ V15; Subfamily P: TCRβ V30; Subfamily Q: TCRβ V19; Subfamily R:TCRβ V27; Subfamily S:TCRβ V28; Subfamily T: TCRβ V24; Subfamily U: TCRβ V20; Subfamily V: TCRβ V25; and Subfamily W:TCRβ V29 subfamily. Subfamily members are described in detail herein in the Section titled “TCR beta V (TCRβV)”.

FIGS. 4A-4D show human CD3+ T cells activated by anti-TCR Vβ13.1 antibody (A-H.1) for 6-days. Human CD3+ T cells were isolated using magnetic-bead separation (negative selection) and activated with immobilized (plate-coated) anti-TCR Vβ13.1 (A-H.1) or anti-CD3E (OKT3) antibodies at 100 nM for 6 days. FIG. 4A shows two scatter plots (left: activated with OKT3; and right: activated with A-H.1) of expanded T cells assessed for TCR Vβ13.1 surface expression using anti-TCR Vβ13.1 (A-H.1) followed by a secondary fluorochrome-conjugated antibody for flow cytometry analysis. FIG. 4B shows two scatter plots (left: activated with OKT3; and right: activated with A-H.1) of expanded T cells assessed for TCR Vβ13.1 surface expression using anti-TCR Vβ13.1 (A-H.1) followed by a secondary fluorochrome-conjugated antibody for flow cytometry analysis, continued from FIG. 4A. FIG. 4C shows percentage (%) of TCR Vβ13.1 positive T cells activated by anti-TCR Vβ13.1 (A-H.1) or anti-CD3e (OKT3) plotted against total T cells (CD3+). FIG. 4D shows relative cell count acquired by counting the number of events in each T cell subset gate (CD3 or TCR Vβ13.1) for 20 seconds at a constant rate of 60 μl/min. Data shown as mean value from 3 donors.

FIGS. 5A-5B show cytolytic activity of human CD3+ T cells activated by anti-TCR Vβ13.1 antibody (A-H.1) against transformed cell line RPMI 8226. FIG. 5A depicts target cell lysis of human CD3+ T cells activated with A-H.1 or OKT3. Human CD3+ T cells were isolated using magnetic-bead separation (negative selection) and activated with immobilized (plate-coated) A-H.1 or OKT3 at the indicated concentrations for 4 days prior to co-culture with RPMI 8226 cells at a (E:T) ratio of 5:1 for 2 days. Samples were next analyzed for cell lysis of RPMI 8226 cells by FACS staining for CFSE/CD138-labeled, and membrane-impermeable DNA dyes (DRAQ7) using flow cytometry analysis. FIG. 5B shows target cell lysis of human CD3+ T cells activated with A-H.1 or OKT3 incubated with RPMI-8226 at a (E:T) ratio of 5:1 for 6 days followed by cell lysis analysis of RPMI 8226 cells as described above. Percentage (%) target cell lysis was determined by normalizing to basal target cell lysis (i.e. without antibody treatment) using the following formula, [(x−basal)/(100%−basal), where x is cell lysis of sample]. Data shown is a representative of n=1 donor.

FIGS. 6A-6B show IFNg production by human PBMCs activated with the indicated antibodies. Human PBMCs were isolated from whole blood from the indicated number of donors, followed by solid-phase (plate-coated) stimulation with the indicated antibodies at 100 Nm. Supernatant was collected on Days 1, 2, 3, 5, or 6. FIG. 6A is a graph comparing the production of IFNg in human PBMCs activated with the antibodies indicated activated with anti-TCR Vβ13.1 antibodies (A-H.1 or A-H.2) or anti-CD3e antibodies (OKT3 or SP34-2) on Day 1, 2, 3, 5, or 6 post-activation. FIG. 6B shows IFNg production in human PBMCs activated with the antibodies indicated activated with the indicated anti-TCR Vβ13.1 antibodies or anti-CD3e antibody (OKT3) on Day 1, 2, 3, 5, or 6 post-activation.

FIGS. 7A-7B show IL-2 production by human PBMCs activated with the indicated antibodies. A similar experimental setup as described for FIGS. 6A-6B was used.

FIGS. 8A-8B show IL-6 production by human PBMCs activated with the indicated antibodies. A similar experimental setup as described for FIGS. 6A-6B was used.

FIGS. 9A-9B show TNF-alpha production by human PBMCs activated with the indicated antibodies. A similar experimental setup as described for FIGS. 6A-6B was used.

FIGS. 10A-10B show IL-1beta production by human PBMCs activated with the indicated antibodies. A similar experimental setup as described for FIGS. 6A-6B was used.

FIGS. 11A-11B are graphs showing delayed kinetics of IFNg secretion in human PMBCs activated by anti-TCR Vβ13.1 antibody A-H.1 when compared to PBMCs activated by anti-CD3e antibody OKT3. FIG. 11A shows IFNg secretion data from 4 donors. FIG. 11B shows IFNg secretion data from 4 additional donors. Data shown is representative of n=8 donors.

FIG. 12 depicts increased CD8+ TSCM and Temra T cell subsets in human PBMCs activated by anti-TCR Vβ13.1 antibodies (A-H.1 or A-H.2) compared to PBMCs activated by anti-CD3e antibodies (OKT3 or SP34-2).

FIGS. 13A-13F show characterization of an anti-TCRVb antibody. FIG. 13A is a graph depicting proliferation of T cells activated with anti-CD3 (OKT3) antibody or anti-TCRVb antibody. FIG. 13B shows selective expansion of CD45RA+ effector memory CD8+ and CD4+ T cells (TEMRA) cells with anti-TCRVb antibodies. Tn=naïve T cell; Tscm=stem cell memory T cell; Tcm=central memory T cell; Tem=effector memory T cell; Temra=effector memory CD45RA+ T cell. FIG. 13C is a graph showing IFN-g secretion by PBMCs stimulated with an anti-TCRVb antibody, or anti-CD3 antibodies. FIG. 13D shows target cell lysis by T cells stimulated with an anti-TCRVb antibody, or anti-CD3 antibodies. Cells were stimulated for 4 days followed by 2 days incubation with multiple myeloma target cells for assessment of cell killing. FIG. 13E is a graph showing perforin secretion by T cells stimulated with an anti-TCRVb antibody, or an anti-CD3 antibody. Perforin was analyzed by FACS staining in TCRVB-positive and TCRVB-negative T cells in PBMCs after 5 days of stimulation with 100 ng/ml plate-bound antibody. FIG. 13F is a graph showing Granzyme B by T cells stimulated with an anti-TCRVb antibody, or an anti-CD3 antibody. Granzyme B was analyzed by FACS staining in TCRVB-positive and TCRVB-negative T cells in PBMCs after 5 days of stimulation with 100 ng/ml plate-bound antibody.

FIGS. 14A-14C show production of IL-2 and IL-15 and expansion of human NK cells by stimulation of PBMCs with anti-TCRVb antibody for 6 days at a dose of 100 nM. FIG. 14A shows secretion of IL-2 or IL-15 in T cells stimulated with an anti-TCRVb antibody, or anti-CD3 antibodies. FIG. 14B depicts flow cytometry dot plots showing NKp46 staining vs CD56 antibody staining in cells stimulated with an anti-TCRVb antibody or an anti-CD3 antibody or a control sample. FIG. 14C depicts flow cytometry dot plots showing NKp46 staining vs CD56 antibody staining in cells stimulated with an anti-TCRVb antibody or an anti-CD3 antibody or a control sample, continued from FIG. 14B.

FIGS. 15A-15C show secretion of cytokines in PBMCs stimulated with an anti-TCRVb antibody, or anti-CD3 antibodies. FIG. 15A, IL-6; FIG. 15B, IL-10; FIG. 15C, TNFα

FIGS. 16A-16B show killing of MM cells by dual targeting BCMA-TCRvb antibody molecules. FIG. 16A shows in vitro killing by one of the following dual-targeting antibody molecules: BCMA-TCRVb (Molecule I), BCMA-CD3, or Control-TCRVb; or an isotype control.

FIG. 16B shows in vivo killing of MM cells by a dual-targeting BCM-TCRVb antibody (Molecule I).

FIG. 17 shows lysis of MM target cells with a dual targeting antibody (Molecule E) which recognized FcRH5 on one arm and TCRVb on the other arm.

FIGS. 18A-18B demonstrate cytokine production from human PBMCs activated by anti-TCR Vβ8a antibodies (B-H.1) when compared to those activated by anti-CD3E antibodies (OKT3 or SP34-2). FIG. 18A shows that human PBMCs activated by anti-TCR Vβ8a antibodies (B-H.1) produce similar or reduced levels of IFNγ. FIG. 18B shows human PBMCs activated by anti-TCR Vβ8a antibodies (B-H.1) produce higher levels of IL-2 when compared to those activated by anti-CDR antibodies (OKT3 or SP34-2). Data shown is representative of n=6 donors.

FIGS. 19A-19C demonstrate cytokine production from human PBMCs activated by anti-TCR Vβ8a antibodies (B-H.1). Human PBMCs activated by anti-TCR Vβ8a antibodies (B-H.1) do not significantly produce IL-6 (FIG. 19A), IL1b (FIG. 19B), and less TNFa (FIG. 19C), when compared to PBMCs activated by anti-CD3E antibodies (OKT3 or SP34-2). Data shown is representative of n=6 donors.

FIGS. 20A-20E demonstrate cytokine production from human PBMCs activated by anti-TCRβV Antibody D antibody compared to control anti-CD3e antibody (OKT3). FIG. 20A shows that human PBMCs activated by anti-TCRβV Antibody D antibody produce similar or reduced levels of IFNγ. FIG. 20B shows human PBMCs activated by anti-TCRβV Antibody D antibody produce higher levels of IL-2 when compared to those activated by anti-CD3E antibodies (OKT3). Human PBMCs activated by anti-TCRβV Antibody D antibody do not significantly produce IL-1beta (FIG. 20C), IL-6, (FIG. 20D), or TNFalpha (FIG. 20E). Data shown is representative of n=4 donors.

FIGS. 21A-21B demonstrate cytokine production from human PBMCs activated by anti-TCR V135 antibody (Antibody E). FIG. 21A shows that human PBMCs activated by anti-TCR Vβ5 antibody produce similar or reduced levels of IFNγ compared to PBMCS activated by anti-CD3ϵ antibodies (OKT3 or SP34-2). FIG. 21B shows human PBMCs activated by the anti-TCR Vβ5 1 antibody produce higher levels of IL-2 when compared to those activated by anti-CD3ϵ antibodies (OKT3 or SP34-2). Data shown is representative of n=4 donors.

FIGS. 22A-22D demonstrate cytokine production from human PBMCs activated by an anti-TCR V135 antibody (Antibody E). Human PBMCs activated by anti-TCR V135 antibody do not significantly produce IL-1beta (FIG. 22A), IL-6, (FIG. 22B), TNFalpha (FIG. 22C), or IL-10 (FIG. 22D) as compared to PBMCs activated by anti-CD3E antibodies (OKT3 or SP34-2). Data shown is representative of n=4 donors.

FIGS. 23A-23F demonstrate cytokine production from human PBMCs activated by a dual targeting (bispecific molecule) comprising an anti-TCRβV binding moiety and a BCMA binding moiety. FIG. 23A shows that human PBMCs activated by the bispecific molecule produce similar or reduced levels of IFNγ as PBMCS activated by anti-CD3E antibodies (OKT3). FIG. 23B shows human PBMCs activated by the bispecific molecule produce higher levels of IL-2 when compared to PBMCs activated by anti-CD3E antibodies (OKT3). Human PBMCs activated by the bispecific molecule do not significantly produce IL-1beta (FIG. 23C), IL-6, (FIG. 23D), TNFalpha (FIG. 23E), or IL-10 (FIG. 23F). Data shown is representative of n=3 donors.

FIGS. 24A-24B show the structure and sequence of eight TCRβV proteins from seven different subfamilies: TCRβV6 subfamily (TCRβV6-5 and TCRβV6-4 are shown), TCRβV28 subfamily, TCRβV19 subfamily, TCRβV9 subfamily, TCRβV5 subfamily, TCRβV20 subfamily and TCRβV12 subfamily. FIG. 24A shows the structural alignment of the different TCRβV proteins. The circled area represents the outward facing region comprising the proposed binding site for the anti-TCRβV antibodies disclosed herein. FIG. 24B shows the amino acid sequence alignment of the proteins shown in FIG. 24A (SEQ ID NOS: 3449-3456, respectively, in order of appearance). The various TCRβV proteins (from 7 different TCRβV subfamilies) have diverse sequences but share a conserved (similar) structure and function.

FIGS. 25A-25J show cytokine or chemokine secretion of PBMCs activated with anti-TCRVb antibodies (A-H.1, B-H.1), a bispecific molecule comprising an anti-TCRVb antibody (Molecule H), control isotype (122) or anti-CD3e antibody (OKT3). Data shown is representative of n=2 donors and representative of 2 independent experiments. FIG. 25A, IFNγ; FIG. 25B, IL-2; FIG. 25C, IL-1β; FIG. 25D, IL-6; FIG. 25E, IL-10; FIG. 25F, IL-4; FIG. 25G, TNFα; FIG. 2511, IL-12p70; FIG. 251, IL-13; FIG. 25J, IL-8.

FIGS. 26A-26H show cytokine or chemokine secretion of PBMCs activated with anti-TCRVb antibodies (A-H.1, B-H.1), a bispecific molecule comprising an anti-TCRVb antibody (Molecule H), control isotype (122) or anti-CD3e antibody (OKT3). Data shown is representative of n=2 donors and representative of 2 independent experiments. FIG. 26A, Eotaxin; FIG. 26B, Eotaxin-3; FIG. 26C, IL-8 (HA); FIG. 26D, IP-10; FIG. 26E, MCP-1; FIG. 26F, MCP-4; FIG. 26G, MDC; FIG. 2611, MIP-1a.

FIGS. 27A-27L show cytokine or chemokine secretion of PBMCs activated with anti-TCRVb antibodies (A-H.1, B-H.1), a bispecific molecule comprising an anti-TCRVb antibody (Molecule H), control isotype (122) or anti-CD3e antibody (OKT3). Data shown is representative of n=2 donors and representative of 2 independent experiments. FIG. 27A, MIP-1B; FIG. 27B, TARC; FIG. 27C, GM-CSF; FIG. 27D, IL-12-23p40; FIG. 27E, IL-15; FIG. 27F, IL-16; FIG. 27G, IL-17a; FIG. 2711, IL-1a; FIG. 271, IL-5; FIG. 27J, IL-7; FIG. 27K, TNF-β; FIG. 27L, VEGF.

FIG. 28 is a graph depicting mean tumor volume in NOD/SCID/IL-2Rγnull (NSG) mice engrafted with Raji-luc cells at days 10 to 28. The Star denotes PBMC implantation. Open triangles denote antibody treatment with the indicated antibodies.

FIGS. 29A-29B depicting Mean tumor burden (Total Flux) in NOD/SCID/IL-2Rγnull (NSG) mice engrafted with cancer cells and treated with the indicated antibody. NSG mice were implanted with PBMCs on Day 1 followed by injection with cancer cells on Day 7 (Raji-luc in FIG. 29A; K562-Luc control in FIG. 29B). Antibody treatment with the indicated antibodies began on Day 16. FIG. 29A shows mean tumor burden at days 16 to 37 in NOD/SCID/IL-2Rγnull (NSG) mice engrafted with Raji-luc cells. FIG. 29B shows mean tumor burden (Total Flux) at days 16 to 30 in animals engrafted with K562-luc cells.

FIG. 30 is a graph depicting Mean tumor burden (Total Flux) mean tumor volume in NOD/SCID/IL-2Rγnull (NSG) mice engrafted with RPMI-8226 cells. The RPMI-8226 cells were engrafted on Day 1. On Day 11, PBMCs were implanted into the mice and antibody treatment began on Day 17.

FIGS. 31A-31B are graphs showing % target cell lysis at different antibody concentrations. FIG. 31A shows data generated using anti-TCR Vβ13.1/anti-CD19 (Molecule F), anti-CD3/anti-CD19, and anti-TCR Vβ13.1 (A-H.1). FIG. 31B shows data generated using anti-TCR Vβ13.1/anti-BCMA (Molecule G), anti-CD3/anti-BCMA, and anti-TCR Vβ13.1 (A-H.1).

FIGS. 32A-32F are graphs showing cytokine secretion stimulated by anti-TCR Vβ/anti-BCMA (Molecule H) or anti-CD3 (OKT3) at Days 1, 2, 3, and 5. Cytokines examined include: IFNγ (FIG. 32A), IL-2 (FIG. 32B), IL-1β (FIG. 32C), IL-6 (FIG. 32D), IL-10 (FIG. 32E), and TNFα (FIG. 32F).

FIGS. 33A-33F are graphs showing cytokine secretion stimulated by anti-TRBC1 (Antibody F) or anti-CD3 (OKT3) at Days 2 and 5. Cytokines examined include: IFNγ (FIG. 33A), IL-2 (FIG. 33B), IL-1(3 (FIG. 33C), IL-6 (FIG. 33D), IL-10 (FIG. 33E), and TNFα (FIG. 33F).

FIGS. 34A-34B are FACS plots showing the expansion of TCRvb 6-5+ T cells over 8 days using anti-TCRvb 6-5 v1. FIG. 34A, Day 0, Day 1, Day 2; FIG. 34B, Day 4, Day 6, Day 8.

FIG. 35 is a bar graph showing the expansion of TCRvb 6-5+ CD4+ T cells and TCRvb 6-5+ CD8+ T cells over 8 days using the anti-CD3ε antibody OKT3 (100 nM).

FIG. 36 is a bar graph showing the expansion of TCRvb 6-5+ CD4+ T cells and TCRvb 6-5+ CD8+ T cells over 8 days using the anti-TCRvb 6-5 v1 antibody (100 nM).

FIG. 37 is a FACS plot showing the showing the expansion of TCRvb 6-5+ T cells over 8 days using anti-TCRvb 6-5 v1 or the anti-CD3ε antibody OKT3.

FIG. 38A is a bar graph showing the percentage of TCRβV 6-5+ T cells in PBMC cultures after 8 days of culture with the indicated antibody. Data for 5 replicates are shown. FIG. 38B is a bar graph showing the percentage of TCRβV 6-5+ T cells in purified T cell cultures after 8 days of culture with the indicated antibody. Data for 5 replicates are shown.

FIG. 39A is a bar graph showing the relative count of TCRβV 6-5+ T cells in PBMC culture after 8 days of culture with the indicated antibody. FIG. 39B is a bar graph showing the relative count of TCRβV 6-5+ T cells in PBMC culture after 8 days of culture with the indicated antibody.

FIG. 40A is a bar graph showing the relative count of TCRβV 6-5+ T cells in a purified T cell culture after 8 days of culture with the indicated antibody. FIG. 40B is a bar graph showing the relative count of TCRβV 6-5+ T cells in a purified T cell culture after 8 days of culture with the indicated antibody.

FIG. 41 is a line graph showing the total CD3+ T cell count (fold increase) after 8 days of T cell culture with either the anti-CD3ε antibody OKT3 or the anti-TCRvb 6-5 v1 antibody.

FIG. 42 is a series of line graphs showing the kinetics of target cells by TCRβV 6-5 v1 activated T cells or anti-CD3ε (OKT3) activated T cells. T cells from three different donors were utilized (donor 6769, donor 9880, donor 5411).

FIG. 43A is a scatter plot showing the percent of target cell lysis by T cells by TCRβV 6-5 v1 activated T cells or anti-CD3ε (OKT3) activated T cells without T cell pre activation. The data is presented at day 6 of co-culture between target cells and effector T cells. FIG. 43B is a scatter plot showing the percent of target cell lysis by T cells by TCRβV 6-5 v1 activated T cells or anti-CD3ε (OKT3) activated T cells with 4 days of T cell pre activation. The data is presented at day 2 of co-culture between target cells and effector T cells (after 4 days of T cell pre-activation).

FIG. 44 is a scatter plot showing the percent of target cell lysis by T cells by TCRβV 6-5 v1 activated T cells or anti-CD3ε (OKT3) activated T cells with 4 days of T cell pre activation. The data is presented at day 2 of co-culture between target cells and effector T cells (after 4 days of T cell pre-activation).

FIG. 45 is a bar graph showing target cell lysis by T cells by TCRβV 6-5 v1 activated T cells or anti-CD3ε (OKT3) activated T cells (100 nM each antibody). The data includes seven replicates of each experimental condition.

FIG. 46 is a series of FACS plots that show the cell surface expression of CD3c on CD4+ TCRβV 6-5 or CD4+ TCRβV 6-5+ T cells activated with either SP34-2 (anti-CD3ε antibody) or anti-TCRβV 6-5 v1 (anti-TCRβV 6-5 antibody) at days 0, 1, 2, 4, 6, or 8 post antibody activation.

FIG. 47 is a series of FACS plots that show the cell surface expression of CD3c on CD8+ TCRβV 6-5 or CD8+ TCRβV 6-5+ T cells activated with either SP34-2 (anti-CD3ε antibody) or anti-TCRβV 6-5 v1 (anti-TCRβV 6-5 antibody) at days 0, 1, 2, 4, 6, or 8 post antibody activation.

FIG. 48 is a series of FACS plots that show the cell surface expression of TCRβV on CD4+ TCRβV 6-5+ or CD4+ TCRβV 6-5+ T cells activated with either SP34-2 (anti-CDR antibody) or anti-TCRβV 6-5 v1 (anti-TCRβV 6-5 antibody) at days 0, 1, 2, 4, 6, or 8 post antibody activation.

FIG. 49 is a series of FACS plots that show the cell surface expression of TCRβV on CD8+ TCRβV 6-5+ or CD8+ TCRβV 6-5+ T cells activated with either SP34-2 (anti-CDR antibody) or anti-TCRβV 6-5 v1 (anti-TCRβV 6-5 antibody) at days 0, 1, 2, 4, 6, or 8 post antibody activation.

FIG. 50A shows FACS plot of TCRβV 6-5+ cynomolgus T cell expansion either unstimulated (left) or stimulated with anti-TCRβV 6-5 v1 (right) 7 days post activation of cynomolgus PBMCs. PBMCs from Donor DW8N (fresh PBMC sample, male, age 8, weight 7.9 kgs) were used. FIG. 50B shows FACS plot of TCRβV 6-5+ cynomolgus T cell expansion either unstimulated (left) or stimulated with anti-TCRβV 6-5 v1 (right) 7 days post activation of cynomolgus PBMCs. PBMCs from Donor G709 (cryopreserved sample, male, age 6, weight 4.7 kgs) were used.

FIG. 51 shows FACS plot and corresponding microscopy images of TCRβV 6-5+ cynomolgus T cell expansion either unstimulated (left), stimulated with SP34-2 (anti-CD3ε antibody) (middle); or stimulated with anti-TCRβV 6-5 v1 (right) post activation of cryopreserved donor DW8N cynomolgus PBMCs. The microscopy images show the cell cluster formation (indicated by circles).

FIG. 52 shows a schematic of FACS plot showing the FACS gating/staining of PBMCs prior γδ T cell purification.

FIG. 53 shows a schematic of FACS plot showing the FACS gating/staining of purified γδ T cell population.

FIG. 54A show activation of purified γδ T cell population with anti-CD3ε antibody (SP34-2) (left) or anti-TCRβV antibody (anti-TCRβV 6-5 v1) (right). FIG. 54B show activation of purified γδ T cell population with anti-CD3ε antibody (SP34-2) (left) or anti-TCRβV antibody (anti-TCRβV 6-5 v1) (right), continued from FIG. 54A.

FIG. 55A shows the release of IFNγ from purified γδ T cell populations activated with anti-CD3ε antibody (SP34-2), anti-TCRβV antibody (anti-TCRβV 6-5 v1), or unstimulated. FIG. 55B shows the release of TNFα from purified γδ T cell populations activated with anti-CD3ε antibody (SP34-2), anti-TCRβV antibody (anti-TCRβV 6-5 v1), or unstimulated. FIG. 55C shows the release of IL-2 from purified γδ T cell populations activated with anti-CD3ε antibody (SP34-2), anti-TCRβV antibody (anti-TCRβV 6-5 v1), or unstimulated. FIG. 55D shows the release of IL-17A from purified γδ T cell populations activated with anti-CD3ε antibody (SP34-2), anti-TCRβV antibody (anti-TCRβV 6-5 v1), or unstimulated. FIG. 55E shows the release of IL-1α from purified γδ T cell populations activated with anti-CD3ε antibody (SP34-2), anti-TCRβV antibody (anti-TCRβV 6-5 v1), or unstimulated. FIG. 55F shows the release of IL-10 from purified γδ T cell populations activated with anti-CD3ε antibody (SP34-2), anti-TCRβV antibody (anti-TCRβV 6-5 v1), or unstimulated. FIG. 55G shows the release of IL-6 from purified γδ T cell populations activated with anti-CD3ε antibody (SP34-2), anti-TCRβV antibody (anti-TCRβV 6-5 v1), or unstimulated. FIG. 55H shows the release of IL-10 from purified γδ T cell populations activated with anti-CD3ε antibody (SP34-2), anti-TCRβV antibody (anti-TCRβV 6-5 v1), or unstimulated.

FIG. 56A shows the relative representations of all TCR alpha V segments (TRAV group of genes) and their variants (top), all TCR beta V segment 6-5 variants (TRBV6-5 gene) (bottom left), and all TCR beta V segments and variants excluding 6-5 (bottom right). FIG. 56B shows the relative representations of all TCR alpha V segments (TRAV group of genes) and their variants (top), all TCR beta V segment 6-5 variants (TRBV6-5 gene) (bottom left), and all TCR beta V segments and variants excluding 6-5 (bottom right), continued from FIG. 56A.

FIG. 57A is a FACS plot showing phenotypic markers of CD4+ T cells expanded with anti-TCRβV antibody (anti-TCRβV 6-5 v1). Defined phenotypes include TEMRA (top left), Naïve/TSCM (top right), TEM (bottom left), and TCM (bottom right). FIG. 57B is a FACS plot showing phenotypic markers of CD4+ T cells expanded with anti-CD3ε antibody (OKT3). Defined phenotypes include TEMRA (top left), Naïve/TSCM (top right), TEM (bottom left), and TCM (bottom right).

FIG. 58A is a FACS plot showing phenotypic markers of CD8+ T cells expanded with anti-TCRβV antibody (anti-TCRβV 6-5 v1). Defined phenotypes include TEMRA (top left), Naïve/TSCM (top right), TEM (bottom left), and TCM (bottom right). FIG. 58B is a FACS plot showing phenotypic markers of CD8+ T cells expanded with anti-CD3ε antibody (OKT3). Defined phenotypes include TEMRA (top left), Naïve/TSCM (top right), TEM (bottom left), and TCM (bottom right).

FIG. 59A is a bar graph showing the percentage of PD1 expressing CD4+ T cells from T cell cultures activated with anti-TCRβV antibody (anti-TCRβV 6-5 v1), anti-CD3ε antibody (OKT3), or unstimulated. FIG. 59B is a bar graph showing the percentage of PD1 expressing CD8+ T cells from T cell cultures activated with anti-TCRβV antibody (anti-TCRβV 6-5 v1), anti-CD3ε antibody (OKT3), or unstimulated.

FIG. 60A is a bar graph showing the expression of Ki-67 by CD4+ T cells from T cell cultures activated with anti-TCRβV antibody (anti-TCRβV 6-5 v1), anti-CD3ε antibody (OKT3), or unstimulated. FIG. 60B is a bar graph showing the expression of Ki-67 by CD8+ T cells from T cell cultures activated with anti-TCRβV antibody (anti-TCRβV 6-5 v1), anti-CD3ε antibody (OKT3), or unstimulated.

FIG. 61A is a FACS plot showing the percentage of TEMRA-like CD8+ T cells activated using anti-TCRβV antibody (anti-TCRβV 6-5 v1) that express CD57 (18.7%). FIG. 61B is a FACS plot showing the percentage of TEM-like CD8+ T cells activated using anti-CD3ε antibody (OKT3) that express CD57 (46.8%) and the percentage of TCM-like CD8+ T cells activated using anti-CD3ε antibody (OKT3) that express CD57 (18.9%).

FIG. 62 shows a series of FACS plots showing the expression of expression of CD27 and by CD4+ (top) or CD8+ (bottom) T cells from T cell cultures activated with anti-TCRβV antibody (anti-TCRβV 6-5 v1), anti-CD3ε antibody (OKT3), or unstimulated.

FIG. 63 shows a series of FACS plots showing the expression of expression of OX40, 41BB, and ICOS by CD4+ (top) or CD8+ (bottom) T cells from T cell cultures activated with anti-TCRβV antibody (anti-TCRβV 6-5 v1), anti-CD3ε antibody (OKT3), or unstimulated.

FIG. 64A shows a series of FACS plots showing the percentage of CD3+ (CD4 gated) TCRβV 6-5+ T cells 1, 2, 3, 4, 5, 6, and 8 days port activation with BCMA and the anti-TCR antibody anti-TCR Bβ 6-5 v1. FIG. 64B shows a series of FACS plots showing the percentage of CD3+ (CD4 gated) TCRβV 6-5+ T cells 1, 2, 3, 4, 5, 6, and 8 days port activation with BCMA and the anti-TCR Vβ antibody anti-TCR Vβ 6-5 v1, continued from FIG. 64A.

FIG. 65A shows a series of FACS plots showing the percentage of CD4+ T cells expanded using isotype control (IgG1 N297A), anti-TCRβV (anti-TCR VP 6-5 v1), or anti-CD3ε (OKT3) antibodies on day 0 post activation. FIG. 65B shows a series of FACS plots showing the percentage of CD4+ T cells expanded using isotype control (IgG1 N297A), anti-TCRβV (anti-TCR VP 6-5 v1), or anti-CD3ε (OKT3) antibodies on day 1 post activation. FIG. 65C shows a series of FACS plots showing the percentage of CD4+ T cells expanded using isotype control (IgG1 N297A), anti-TCRβV (anti-TCR VP 6-5 v1), or anti-CD3ε (OKT3) antibodies on day 2 post activation. FIG. 65D shows a series of FACS plots showing the percentage of CD4+ T cells expanded using isotype control (IgG1 N297A), anti-TCRβV (anti-TCR VP 6-5 v1), or anti-CD3ε (OKT3) antibodies on day 3 post activation. FIG. 65E shows a series of FACS plots showing the percentage of CD4+ T cells expanded using isotype control (IgG1 N297A), anti-TCRβV (anti-TCR VP 6-5 v1), or anti-CD3ε (OKT3) antibodies on day 4 post activation. FIG. 65F shows a series of FACS plots showing the percentage of CD4+ T cells expanded using isotype control (IgG1 N297A), anti-TCRβV (anti-TCR VP 6-5 v1), or anti-CD3ε (OKT3) antibodies on day 5 post activation. FIG. 65G shows a series of FACS plots showing the percentage of CD4+ T cells expanded using isotype control (IgG1 N297A), anti-TCRβV (anti-TCR VP 6-5 v1), or anti-CD3ε (OKT3) antibodies on day 6 post activation. FIG. 65H shows a series of FACS plots showing the percentage of CD4+ T cells expanded using isotype control (IgG1 N297A), anti-TCRβV (anti-TCR VP 6-5 v1), or anti-CD3ε (OKT3) antibodies on day 8 post activation.

FIG. 66A is a bar graph showing ATP production from glycolysis of T cell cultures activated with the indicated antibodies. FIG. 66B is a bar graph showing ATP production from oxidative phosphorylation of T cell cultures activated with the indicated antibodies.

FIG. 67 is a line graph showing the oxygen consumption rate (OCR) of T cells from about 0 to 75 minutes activated with the indicated antibody.

FIG. 68A shows the oxygen consumption rate (OCR) of T cells activated with the indicated antibody during basal respiration. FIG. 68B shows the oxygen consumption rate (OCR) of T cells activated with the indicated antibody during maximal respiration. FIG. 68C shows the oxygen consumption rate (OCR) of T cells activated with the indicated antibody during spare respiratory capacity. FIG. 68D is a line graph indicates the areas of basal respiration and maximal respiration as shown in FIG. 68A and FIG. 68B, respectively.

FIG. 69A is a bar graph showing ATP production from glycolysis of T cell cultures activated with anti-TCRβV 6-5 v1 and re-stimulated with the indicated antibody. FIG. 69B is a bar graph showing ATP production from oxidative phosphorylation of T cell cultures activated with anti-TCRβV 6-5 v1 and re-stimulated with the indicated antibody.

FIGS. 70A-70G are graphs showing expression of IFNg (FIG. 70A), TNFα (FIG. 70E), IL-1a (FIG. 70B), IL-1b (FIG. 70C), IL-6 (FIG. 70D), IL-10 (FIG. 70F), IL-17A (FIG. 70G) (CRS and neurotoxicity associated cytokines) with BHM1710 (anti TCRVB), a reduced affinity anti CD3 antibody (TB) and the SP34 anti CD3e antibody.

FIG. 71 is a FACS plot showing the percentage of NK cells expanded from T cell cultures activated with the indicated antibody.

FIG. 72 is a bar graph showing the number of NK cells expanded from T cell cultures activated with the indicated antibody.

FIG. 73 shows a series of FACS plots showing NK cell proliferation induced by T cell cultures activated with the indicated antibody.

FIG. 74 is a schematic showing an assay described in Example for determining NK cell mediated lysis of target K562 cells.

FIG. 75 is a bar graph showing the percent target cell lysis mediated by NK cells activated by PBMCs activated with the indicated antibody.

FIG. 76A shows a series of FACS plots showing the proliferation of NK cells from PBMC cultures activated/expanded with the indicated antibody (isotype control or OKT3). PBMCs from three donors (D1, D2, and D3) were analyzed. FIG. 76B shows a series of FACS plots showing the proliferation of NK cells from PBMC cultures activated/expanded with the indicated antibody (isotype control or OKT3). PBMCs from three donors (D1, D2, and D3) were analyzed, continued from FIG. 76A.

FIG. 77A shows a series of FACS plots showing the proliferation of NK cells from PBMC cultures activated/expanded with the indicated antibody (anti-TCRvβ 12-3/4 v1 or anti-TCRvβ12-3/4 v2). PBMCs from three donors (D1, D2, and D3) were analyzed. FIG. 77B shows a series of FACS plots showing the proliferation of NK cells from PBMC cultures activated/expanded with the indicated antibody (anti-TCRvβ 12-3/4 v1 or anti-TCRvβ 12-3/4 v2). PBMCs from three donors (D1, D2, and D3) were analyzed, continued from FIG. 77A.

FIG. 78A shows a series of FACS plots showing the proliferation of NK cells from PBMC cultures activated/expanded with the indicated antibody (anti-TCRvβ 12-3/4 v3 or SP34-2). PBMCs from three donors (D1, D2, and D3) were analyzed. FIG. 78B shows a series of FACS plots showing the proliferation of NK cells from PBMC cultures activated/expanded with the indicated antibody (anti-TCRvβ 12-3/4 v3 or SP34-2). PBMCs from three donors (D1, D2, and D3) were analyzed, continued from FIG. 78A.

FIG. 79 is a bar graph showing the level of secreted IFNγ by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, OKT3, or SP34) and cultured with said antibody for the indicated number of days (1, 3, or 5).

FIG. 80 is a bar graph showing the level of secreted IL-2 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, OKT3, or SP34) and cultured with said antibody for the indicated number of days (1, 3, or 5).

FIG. 81 is a bar graph showing the level of secreted IL-15 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, OKT3, or SP34) and cultured with said antibody for the indicated number of days (1, 3, or 5).

FIG. 82 is a bar graph showing the level of secreted IL-10 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, OKT3, or SP34) and cultured with said antibody for the indicated number of days (1, 3, or 5).

FIG. 83 is a bar graph showing the level of secreted IL-6 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, OKT3, or SP34) and cultured with said antibody for the indicated number of days (1, 3, or 5).

FIG. 84 is a bar graph showing the level of secreted IL-10 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, OKT3, or SP34) and cultured with said antibody for the indicated number of days (1, 3, or 5).

FIG. 85 is a bar graph showing the level of the indicated cytokine secreted by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 or SP34). The data includes use of 17 individual PBMC donors.

FIG. 86A is a bar graph showing the level of secreted IFNγ by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 or OKT3) and cultured with said antibody for the indicated number of days (1, 2, 3, 5, or 6). FIG. 86B is a bar graph showing the level of secreted IL-10 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 or OKT3) and cultured with said antibody for the indicated number of days (1, 2, 3, 5, or 6).

FIG. 86C is a bar graph showing the level of secreted IL-4 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 or OKT3) and cultured with said antibody for the indicated number of days (1, 2, 3, 5, or 6). FIG. 86D is a bar graph showing the level of secreted IL-6 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 or OKT3) and cultured with said antibody for the indicated number of days (1, 2, 3, 5, or 6). FIG. 86E is a bar graph showing the level of secreted IL-10 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 or OKT3) and cultured with said antibody for the indicated number of days (1, 2, 3, 5, or 6). FIG. 86F is a bar graph showing the level of secreted TNFα by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 or OKT3) and cultured with said antibody for the indicated number of days (1, 2, 3, 5, or 6). FIG. 86G is a bar graph showing the level of secreted IL-2 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 or OKT3) and cultured with said antibody for the indicated number of days (1, 2, 3, 5, or 6).

FIG. 87A is a bar graph showing the level of secreted IFNγ by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, anti-TCRβV 6-5 v1, OKT3, SP34-2, or isotype control) and cultured with said antibody for the indicated number of days (1, 2, 3, 5, or 6). FIG. 87B is a bar graph showing the level of secreted IL-10 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, anti-TCRβV 6-5 v1, OKT3, SP34-2, or isotype control) and cultured with said antibody for the indicated number of days (1, 2, 3, 5, or 6). FIG. 87C is a bar graph showing the level of secreted IL-4 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, anti-TCRβV 6-5 v1, OKT3, SP34-2, or isotype control) and cultured with said antibody for the indicated number of days (1, 2, 3, 5, or 6). FIG. 87D is a bar graph showing the level of secreted IL-6 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, anti-TCRβV 6-5 v1, OKT3, SP34-2, or isotype control) and cultured with said antibody for the indicated number of days (1, 2, 3, 5, or 6). FIG. 87E is a bar graph showing the level of secreted IL-10 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, anti-TCRβV 6-5 v1, OKT3, SP34-2, or isotype control) and cultured with said antibody for the indicated number of days (1, 2, 3, 5, or 6). FIG. 87F is a bar graph showing the level of secreted TNFα by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, anti-TCRβV 6-5 v1, OKT3, SP34-2, or isotype control) and cultured with said antibody for the indicated number of days (1, 2, 3, 5, or 6). FIG. 87G is a bar graph showing the level of secreted IL-2 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, anti-TCRβV 6-5 v1, OKT3, SP34-2, or isotype control) and cultured with said antibody for the indicated number of days (1, 2, 3, 5, or 6).

FIG. 88A is a bar graph showing the level of secreted IFNγ by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, anti-TCRβV 6-5 v1, OKT3, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8).

FIG. 88B is a bar graph showing the level of secreted IL-10 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, anti-TCRβV 6-5 v1, OKT3, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 88C is a bar graph showing the level of secreted IL-4 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, anti-TCRβV 6-5 v1, OKT3, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 88D is a bar graph showing the level of secreted IL-6 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, anti-TCRβV 6-5 v1, OKT3, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 88E is a bar graph showing the level of secreted IL-10 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, anti-TCRβV 6-5 v1, OKT3, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8).

FIG. 88F is a bar graph showing the level of secreted TNFα by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, anti-TCRβV 6-5 v1, OKT3, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 88G is a bar graph showing the level of secreted IL-2 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, anti-TCRβV 6-5 v1, OKT3, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8).

FIG. 89A is a bar graph showing the level of secreted IL-17A by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, OKT3, or SP34-2) and cultured with said antibody for the indicated number of days (2, 5, or 7). FIG. 89B is a bar graph showing the level of secreted IL-17A by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, OKT3, or SP34-2) and cultured with said antibody for the indicated number of days (2, 5, or 8). FIG. 89C is a bar graph showing the level of secreted IL-17A by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1, OKT3, or SP34-2) and cultured with said antibody for the indicated number of days (2, 5, or 7). FIG. 89D is a bar graph showing the level of secreted IL-17A by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 or SP34-2) and cultured with said antibody for the indicated number of days (1, 3, 5, or 7).

FIG. 90A is a bar graph showing the level of secreted IFNγ by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90B is a bar graph showing the level of secreted IL-10 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8).

FIG. 90C is a bar graph showing the level of secreted IL-4 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90D is a bar graph showing the level of secreted IL-6 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90E is a bar graph showing the level of secreted IL-10 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8).

FIG. 90F is a bar graph showing the level of secreted TNFα by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90G is a bar graph showing the level of secreted IL-2 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90H is a bar graph showing the level of secreted IL-12p70 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90I is a bar graph showing the level of secreted IL-13 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90J is a bar graph showing the level of secreted IL-8 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90K is a bar graph showing the level of secreted exotaxin by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8).

FIG. 90L is a bar graph showing the level of secreted exotoxin-3 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90M is a bar graph showing the level of secreted IL-8 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90N is a bar graph showing the level of secreted IP-10 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8).

FIG. 90O is a bar graph showing the level of secreted MCP-1 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90P is a bar graph showing the level of secreted MCP-4 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90Q is a bar graph showing the level of secreted MDC by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90R is a bar graph showing the level of secreted MIP-1a by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90S is a bar graph showing the level of secreted MIP-1b by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90T is a bar graph showing the level of secreted TARC by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8).

FIG. 90U is a bar graph showing the level of secreted GMCSF by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90V is a bar graph showing the level of secreted IL-12-23p40 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90W is a bar graph showing the level of secreted IL-15 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8).

FIG. 90X is a bar graph showing the level of secreted IL-16 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90Y is a bar graph showing the level of secreted IL-17a by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90Z is a bar graph showing the level of secreted IL-1a by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8).

FIG. 90AA is a bar graph showing the level of secreted IL-5 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90BB is a bar graph showing the level of secreted IL-7 by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8). FIG. 90CC is a bar graph showing the level of secreted TNF-β by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8).

FIG. 90DD is a bar graph showing the level of secreted VEGF by T cells activated/expanded with the indicated antibody (isotype control; anti-TCRβV 6-5 v1 with anti-BCMA antibody; anti-TCRβV 6-5 v1; anti-TCRβV 123/4 v1, or SP34-2) and cultured with said antibody for the indicated number of days (1, 2, 3, 4, 5, 6, or 8).

FIG. 91 shows a graphical representation of the relation of sequences between different TCRVB clonotype subfamilies.

FIG. 92A is a bar graph showing the percentage of cytokine release from PBMCs activated/expanded for eight days using the indicated antibody (anti-TCRβV 12-3/4 v1 or SP34-2).

FIG. 92B is a bar graph showing the percentage of cytokine release from PBMCs activated/expanded for eight days using the indicated antibody (anti-TCRβV 5 or SP34-2). FIG. 92C is a bar graph showing the percentage of cytokine release from PBMCs activated/expanded for eight days using the indicated antibody (anti-TCRβV 10 or SP34-2).

FIG. 93A a bar graph showing the level of secreted IFNγ by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6). FIG. 93B a bar graph showing the level of secreted IL-10 by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6). FIG. 93C a bar graph showing the level of secreted IL-17A by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6). FIG. 93D a bar graph showing the level of secreted IL-1α by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6). FIG. 93E a bar graph showing the level of secreted IL-1(3 by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6). FIG. 93F a bar graph showing the level of secreted IL-6 by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6). FIG. 93G a bar graph showing the level of secreted TNFα by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6). FIG. 93H a bar graph showing the level of secreted IL-2 by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6).

FIG. 94 is a bar graph summarizing data from FACS analysis of PBMCs activated/expanded for 6 days using the indicated anti-TCRVβ antibody.

FIG. 95A a bar graph showing the level of secreted IFNγ by T cells activated/expanded with the indicated antibody for the indicated number of days (1, 3, 5, or 7). FIG. 95B a bar graph showing the level of secreted IL-10 by T cells activated/expanded with the indicated antibody for the indicated number of days (1, 3, 5, or 7). FIG. 95C a bar graph showing the level of secreted IL-17A by T cells activated/expanded with the indicated antibody for the indicated number of days (1, 3, 5, or 7). FIG. 95D a bar graph showing the level of secreted IL-1α by T cells activated/expanded with the indicated antibody for the indicated number of days (1, 3, 5, or 7). FIG. 95E a bar graph showing the level of secreted IL-1(3 by T cells activated/expanded with the indicated antibody for the indicated number of days (1, 3, 5, or 7). FIG. 95F a bar graph showing the level of secreted IL-6 by T cells activated/expanded with the indicated antibody for the indicated number of days (1, 3, 5, or 7). FIG. 95G a bar graph showing the level of secreted IL-4 by T cells activated/expanded with the indicated antibody for the indicated number of days (1, 3, 5, or 7). FIG. 95H a bar graph showing the level of secreted IL-2 by T cells activated/expanded with the indicated antibody for the indicated number of days (1, 3, 5, or 7).

FIG. 96 is a bar graph summarizing data from FACS analysis of PBMCs activated/expanded for 7 days using the indicated anti-TCRVβ antibody.

FIG. 97A is a bar graph showing the level of secreted IFNγ by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6). FIG. 97B a bar graph showing the level of secreted IL-10 by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6). FIG. 97C a bar graph showing the level of secreted IL-17A by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6). FIG. 97D a bar graph showing the level of secreted IL-1α by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6). FIG. 97E a bar graph showing the level of secreted IL-1(3 by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6). FIG. 97F a bar graph showing the level of secreted IL-6 by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6). FIG. 97G a bar graph showing the level of secreted IL-4 by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6). FIG. 97H a bar graph showing the level of secreted TNFα by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6). FIG. 97I a bar graph showing the level of secreted IL-2 by T cells activated/expanded with the indicated antibody for the indicated number of days (3 or 6).

FIG. 98A is a bar graph showing the level of secreted IFN-γ by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 (plate coated), anti-CD3ε (plate coated), anti-TCRβV 6-5 v1 (in solution), or anti-CD3ε (in solution) and cultured with said antibody for the indicated number of days (1, 3, 5, or 7). FIG. 98B is a bar graph showing the level of secreted IFN-γ by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 (plate coated), anti-CD3ε (plate coated), anti-TCRβV 6-5 v1 (in solution), or anti-CD3ε (in solution) and cultured with said antibody for the indicated number of days (1, 3, 5, or 7). FIG. 98C is a bar graph showing the level of secreted IL-1b by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 (plate coated), anti-CD3ε (plate coated), anti-TCRβV 6-5 v1 (in solution), or anti-CD3ε (in solution) and cultured with said antibody for the indicated number of days (1, 3, 5, or 7). FIG. 98D is a bar graph showing the level of secreted IL-6 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 (plate coated), anti-CD3ε (plate coated), anti-TCRβV 6-5 v1 (in solution), or anti-CD3ε (in solution) and cultured with said antibody for the indicated number of days (1, 3, 5, or 7). FIG. 98E is a bar graph showing the level of secreted IL-10 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 (plate coated), anti-CD3ε (plate coated), anti-TCRβV 6-5 v1 (in solution), or anti-CD3ε (in solution) and cultured with said antibody for the indicated number of days (1, 3, 5, or 7). FIG. 98F is a bar graph showing the level of secreted IL-15 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 (plate coated), anti-CD3 (plate coated), anti-TCRβV 6-5 v1 (in solution), or anti-CD3c (in solution) and cultured with said antibody for the indicated number of days (1, 3, 5, or 7).

FIG. 98G is a bar graph showing the level of secreted IL-17A by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 (plate coated), anti-CD3ε (plate coated), anti-TCRβV 6-5 v1 (in solution), or anti-CD3ε (in solution) and cultured with said antibody for the indicated number of days (1, 3, 5, or 7). FIG. 98H is a bar graph showing the level of secreted IL-1a by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 (plate coated), anti-CD3ε (plate coated), anti-TCRβV 6-5 v1 (in solution), or anti-CD3ε (in solution) and cultured with said antibody for the indicated number of days (1, 3, 5, or 7). FIG. 98I is a bar graph showing the level of secreted IL-1b by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 (plate coated), anti-CD3ε (plate coated), anti-TCRβV 6-5 v1 (in solution), or anti-CD3ε (in solution) and cultured with said antibody for the indicated number of days (1, 3, 5, or 7). FIG. 98J is a bar graph showing the level of secreted IL-2 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 (plate coated), anti-CD3ε (plate coated), anti-TCRβV 6-5 v1 (in solution), or anti-CD3ε (in solution) and cultured with said antibody for the indicated number of days (1, 3, 5, or 7). FIG. 98K is a bar graph showing the level of secreted IL-4 by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 (plate coated), anti-CD3ε (plate coated), anti-TCRβV 6-5 v1 (in solution), or anti-CD3ε (in solution) and cultured with said antibody for the indicated number of days (1, 3, 5, or 7). FIG. 98L is a bar graph showing the level of secreted TNF-α by T cells activated/expanded with the indicated antibody (anti-TCRβV 6-5 v1 (plate coated), anti-CD3ε (plate coated), anti-TCRβV 6-5 v1 (in solution), or anti-CD3ε (in solution) and cultured with said antibody for the indicated number of days (1, 3, 5, or 7).

FIG. 99 is a FACS plot showing the showing the ability of MH3-2 to bind PBMCs from one of two donors when the PBMCs are either preincubated with TM23 or not (MH3-2 Alone).

FIG. 100A is a FACS plot showing the ability of MH3-2 to bind PBMCs from one of two donors when the PBMCs are either preincubated with TM23 or not (MH3-2 Alone). FIG. 100B is a FACS plot showing the ability of MH3-2 to bind PBMCs from one of two donors when the PBMCs are either preincubated with TM23 or not (MH3-2 Alone), continued from FIG. 100A.

FIG. 101A is a bar graph showing the polyfunctional strength index (PSI) of PBMC CD4+ T cells, CD4+ T cells expanded with anti-CD3 antibody, (CD3 Expanded T cells), and CD4+ T cells expanded with anti-TCRVβ 6-5 antibody (Drug Expanded T cells). The Effector mediators are Granzyme B, IFNγ, MIP-1α, perforin, TNFα, and TNFβ. The Stimulatory mediators are IL-5. The Chemoattractive mediators are MIP-1b. FIG. 101B is a bar graph showing the polyfunctional strength index (PSI) of PBMC CD8+ T cells, CD8+ T cells expanded with anti-CD3 antibody, (CD3 Expanded T cells), and CD8+ T cells expanded with anti-TCRVβ 6-5 antibody (Drug Expanded T cells). The Effector mediators are Granzyme B, IFNγ, MIP-1α, perforin, and TNFβ. The Chemoattractive mediators are MIP-1b and RANTES.

FIGS. 102A-102C show binding of a CD19×TCRvβ bispecific molecule to a TCR molecule. FIG. 102A is a schematic of the bispecific molecule used in this study. FIG. 102B is a graph showing the binding of a CD19×TCRvβ bispecific molecule to soluble TCR. FIG. 102C is a graph showing binding of a CD19×TCRvβ bispecific molecule to TCR expressed on Jurkat cells.

FIGS. 103A-103D show the characterization of a murine CD19×TCRvβ13-2/3 (2×2) bispecific molecule. FIG. 103A is a schematic of the bispecific molecule used in this study. FIG. 103B is a graph showing the binding kinetics of murine CD19×TCRvβ13-2/3. FIG. 103C are dot plots showing the expansion of TCRVB+ T cells following a 6 day incubation with murine CD19×TCRvβ13-2/3. FIG. 103D is a graph showing the relative count of splenic B cells after a 6 day in vitro incubation with murine CD19×TCRvβ13-2/3 bispecific antibody.

FIG. 104 are graphs showing the level of B cells in the blood or spleen of animals treated with 0.1 mg per kg or 1 mg per kg of a murine CD19×TCRvβ13-2/3 bispecific antibody.

FIGS. 105A-105B are graphs showing the level of NK cells (FIG. 105A) or T cells (FIG. 105B) in the blood or spleen of animals treated with 0.1 mg per kg or 1 mg per kg of a murine CD19×TCRvβ13-2/3 bispecific antibody.

FIGS. 106A-106F show expansion of TCRVB+ T cells and lysis of target cells with a CD19×TCRvβ bispecific molecule. FIG. 106A is a schematic of the bispecific molecule used in this study. FIG. 106B is a graph showing target cell lysis by pre-expanded TCRVB+ T cells or CD3+ expanded pan T cells. FIG. 106C shows depletion of purified B cells by purified T cells treated with a CD19×TCRvβ bispecific molecule. FIG. 106D shows depletion of purified B cells by purified T cells treated with a CD19×CD3 bispecific molecule. FIG. 106E shows depletion of B cells in a PBMC preparation treated with a CD19×TCRvβ bispecific molecule. FIG. 106F shows depletion of B cells in a PBMC preparation treated with a CD19×CD3 bispecific molecule.

FIGS. 107A-107D are graphs showing the expression of various cytokines from PBMCs treated with a CD19×CD3 bispecific molecule (FIGS. 107A and 107C (continued from FIG. 107A)) or a CD19×TCRVB 6-5 bispecific molecule (FIGS. 107B and 107D (continued from FIG. 107B)).

FIGS. 108A-108C show a CD19×TCRvβ6-5 (2×2) pharmacokinetic (PK) profile and dosing strategy. FIG. 108A is a schematic of the experimental design. FIG. 108B is a graph showing the concentration of CD19×TCRβ 6-5 at the indicated timepoints after treatment. FIG. 108C shows the detection reagents used to detect CD19×TCRβ 6-5.

FIGS. 109A-109D depicts Table 9 showing the alignment of TCRBV amino acid sequences (SEQ ID NOs: 3457-3516, respectively, in order of appearance). FIG. 109A showing the alignment of TCRBV amino acid sequences (SEQ ID NOs: 3457-3516, respectively, in order of appearance). FIG. 109B showing the alignment of TCRBV amino acid sequences (SEQ ID NOs: 3457-3516, respectively, in order of appearance), continued from FIG. 109A. FIG. 109C showing the alignment of TCRBV amino acid sequences (SEQ ID NOs: 3457-3516, respectively, in order of appearance), continued from FIG. 109B. FIG. 109D showing the alignment of TCRBV amino acid sequences (SEQ ID NOs: 3457-3516, respectively, in order of appearance), continued from FIG. 109C.

FIGS. 110A-110H show the alignment of affinity matured humanized Antibody A-H VL and VH sequences. FIG. 110A shows the alignment of affinity matured humanized Antibody A-H VL sequences (SEQ ID NOS 3377-3389, respectively, in order of appearance), and consensus VL sequence SEQ ID NO: 230, and consensus VL sequence SEQ ID NO: 3289. FIG. 110B shows the alignment of affinity matured humanized Antibody A-H VL sequences (SEQ ID NOS 3377-3389, respectively, in order of appearance), and consensus VL sequence SEQ ID NO: 230, and consensus VL sequence SEQ ID NO: 3289, continued from FIG. 110A. FIG. 110C shows the alignment of affinity matured humanized Antibody A-H VH sequences (SEQ ID NOS 3390-3436, respectively, in order of appearance), and consensus VH sequence SEQ ID NO: 231 and consensus VH sequence SEQ ID NO: 3290. FIG. 110D shows the alignment of affinity matured humanized Antibody A-H VH sequences (SEQ ID NOS 3390-3436, respectively, in order of appearance), and consensus VH sequence SEQ ID NO: 231 and consensus VH sequence SEQ ID NO: 3290, continued from FIG. 110C. FIG. 110E shows the alignment of affinity matured humanized Antibody A-H VH sequences (SEQ ID NOS 3390-3436, respectively, in order of appearance), and consensus VH sequence SEQ ID NO: 231 and consensus VH sequence SEQ ID NO: 3290, continued from FIG. 110D. FIG. 110F shows the alignment of affinity matured humanized Antibody A-H VH sequences (SEQ ID NOS 3390-3436, respectively, in order of appearance), and consensus VH sequence SEQ ID NO: 231 and consensus VH sequence SEQ ID NO: 3290, continued from FIG. 110E. FIG. 110G shows the alignment of affinity matured humanized Antibody A-H VH sequences (SEQ ID NOS 3390-3436, respectively, in order of appearance), and consensus VH sequence SEQ ID NO: 231 and consensus VH sequence SEQ ID NO: 3290, continued from FIG. 110F. FIG. 110H shows the alignment of affinity matured humanized Antibody A-H VH sequences (SEQ ID NOS 3390-3436, respectively, in order of appearance), and consensus VH sequence SEQ ID NO: 231 and consensus VH sequence SEQ ID NO: 3290, continued from FIG. 110G.

DETAILED DESCRIPTION OF THE INVENTION

Current bispecific constructs designed to redirect T cells to promote tumor cell lysis for cancer immunotherapy typically utilize antibody fragments (Fab, scFv, VH, etc.) that are derived from monoclonal antibodies (mAb) directed against the CD3e subunit of the T cell receptor (TCR). However, there are limitations to this approach which may prevent the full realization of the therapeutic potential for such bispecific constructs. Previous studies have shown that even low “activating” doses of anti-CD3e mAb can cause long-term T cell dysfunction and exert immunosuppressive effects. In addition, anti-CD3e mAbs have been associated with side effects that result from massive T cell activation. The large number of activated T cells secrete substantial amounts of cytokines, the most important of which is Interferon gamma (IFNg). This excess amount of IFNg in turn activates macrophages which then overproduce proinflammatory cytokines such as IL-1beta, IL-6, IL-10 and TNF-alpha, causing a “cytokine storm” known as the cytokine release syndrome (CRS) (Shimabukuro-Vornhagen et al., J Immunother Cancer. 2018 Jun. 15; 6(1):56, herein incorporated by reference in its entirety). Thus, the need exists for developing antibodies that are capable of binding and activating only a subset of effector T cells, e.g., to reduce the CRS and/or neurotoxicity (NT).

This invention features molecules targeting the TCRβV chain of TCR and methods thereof. Without wishing to be bound by theory, such molecules are capable of binding, activating, and/or expanding only a subset of T cells, avoiding or reducing CRS and/or NT and minimizing potential immunosuppressive effects of anti-CD3 mAbs.

TCR is a disulfide-linked membrane-anchored heterodimeric protein normally consisting of the highly variable alpha (α) and beta (β) chains expressed as part of a complex with the invariant CD3 chain molecules. TCR on T cells is formed by a heterodimer of one alpha chain and one beta chain. Each alpha or beta chain consists of a constant domain and a highly variable domain classified as the Immunoglobulin superfamily (IgSF) fold. The TCRβV chains can be further classified into 30 subfamilies (TRBV1-30). Despite their high structural and functional homology, the amino acid sequence homology in the TRBV genes is very low. Only 4 amino acids out of ˜95 are identical while 10 additional amino acids are conserved among all subfamilies (see an alignment of TCRBV amino acid sequences in Table 9). Nevertheless, TCRs formed between alpha and beta chains of highly diverse sequences show a remarkable structural homology (FIGS. 24A and 24B) and elicit a similar function, e.g., activation of T cells.

Disclosed herein is the discovery of a novel class of antibodies, i.e., anti-TCRβV antibody molecules disclosed herein, which despite having low sequence similarity (e.g., low sequence identity among the different antibody molecules that recognize different TCRβV subfamilies), recognize a structurally conserved, yet sequence-wise variable, region, e.g., domain, on the TCRβV protein (as denoted by the circled area in FIG. 24A) and have a similar function (e.g., activation of T cells and a similar cytokine profile as described herein). Thus, the anti-TCRβV antibody molecules disclosed herein share a structure-function relationship.

Without wishing to be bound by theory, it is believed that in some embodiments, the anti-TCRβV antibody molecules disclosed herein bind to an outward facing epitope of a TCRβV protein when it is in a complex with a TCRalpha protein, e.g., as denoted by the circled area in FIG. 24A. In some embodiments, the anti-TCRβV antibody molecules disclosed herein recognize (e.g., bind to), a domain (e.g., an epitope) on the TCRβV protein that is: (1) structurally conserved among different TCRβV subfamilies; and (2) has minimal sequence identity among the different TCRβV subfamilies. As shown in Table 9, TCRβV proteins from the different TCRBV subfamilies share minimal sequence similarity. However, as shown in FIG. 24A-B, TCRβV proteins which have minimal sequence similarity, share a similar 3D conformation and structure.

In some embodiments, the anti-TCRβV antibody molecules disclosed herein do not recognize, e.g., bind to, an interface of a TCRβV:TCRalpha complex.

In some embodiments, the anti-TCRβV antibody molecules disclosed herein do not recognize, e.g., bind to, a constant region of a TCRβV protein.

In some embodiments, the anti-TCRβV antibody molecules disclosed herein do not recognize, e.g., bind to, one or more (e.g., all) of a complementarity determining region (e.g., CDR1, CDR2 and/or CDR3) of a TCRβV protein.

This disclosure provides, inter alia, antibody molecules directed to the variable chain of the beta subunit of TCR (TCRβV) which bind and, e.g., activate a subset of T cells. The anti-TCRβV antibody molecules disclosed herein result in lesser or no production of cytokines associated with CRS, e.g., IL-6, IL-1beta, IL-10 and TNF alpha; and enhanced and/or delayed production of IL-2 and IFNg. In some embodiments, the anti-TCRβV antibodies disclosed herein have a cytokine profile, e.g., as described herein, which differs from a cytokine profile of a T cell engager that binds to a receptor or molecule other than a TCRβV region (“a non-TCRβV-binding T cell engager”). In some embodiments, the anti-TCRβV antibodies disclosed herein result in expansion of TCRβV+ T cells, e.g., a subset of memory effector T cells known as TEMRA. Without wishing to be bound by theory, it is believed that in some embodiments, TEMRA cells can promote tumor cell lysis but not CRS. Accordingly, provided herein are methods of making said anti-TCRβV antibody molecules and uses thereof. Also disclosed herein are multispecific molecules, e.g., bispecific molecules comprising said anti-TCRβV antibody molecules. In some embodiments, compositions comprising anti-TCRβV antibody molecules of the present disclosure, can be used, e.g., to: (1) activate and redirect T cells to promote tumor cell lysis for cancer immunotherapy; and/or (2) expand TCRβV+ T cells. In some embodiments, compositions comprising anti-TCRβV antibody molecules as disclosed herein limit the harmful side-effects of CRS and/or NT, e.g., CRS and/or NT associated with anti-CD3e targeting.

In some embodiments, the anti-TCRβV antibody molecule does not bind to TCRβ V12, or binds to TCRβ V12 with an affinity and/or binding specificity that is less than (e.g., less than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the 16G8 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments, the anti-TCRβV antibody molecule binds to TCRβ V12 with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the 16G8 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments, the anti-TCRβV antibody molecule binds to a TCRβV region other than TCRβ V12 (e.g., TCRβV region as described herein, e.g., TCRβ V6 subfamily (e.g., TCRβ V6-5*01) with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the 16G8 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments, the anti-TCRβV antibody molecule does not comprise the CDRs of the Antibody B murine antibody.

In some embodiments, the anti-TCRβV antibody molecule does not bind to TCRβ V5-5*01 or TCRβ V5-1*01, or binds to TCRβ V5-5*01 or TCRβ V5-1*01 with an affinity and/or binding specificity that is less than (e.g., less than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the TM23 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments, the anti-TCRβV antibody molecule binds to TCRβ V5-5*01 or TCRβ V5-1*01 with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the TM23 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments, the anti-TCRβV antibody molecule binds to a TCRβV region other than TCRβ V5-5*01 or TCRβ V5-1*01 (e.g., TCRβV region as described herein, e.g., TCRβ V6 subfamily (e.g., TCRβ V6-5*01) with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the TM23 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments, the anti-TCRβV antibody molecule does not comprise the CDRs of the TM23 murine antibody.

Accordingly, provided herein are, inter alia, anti-TCRβV antibody molecules, multispecific or multifunctional molecules (e.g., multispecific or multifunctional antibody molecules) that comprise anti-TCRβV antibody molecules, nucleic acids encoding the same, methods of producing the aforesaid molecules, pharmaceutical compositions comprising aforesaid molecules, and methods of treating a disease or disorder, e.g., cancer, using the aforesaid molecules. The antibody molecules and pharmaceutical compositions disclosed herein can be used (alone or in combination with other agents or therapeutic modalities) to treat, prevent and/or diagnose disorders and conditions, e.g., cancer, e.g., as described herein.

Definitions

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains.

The term “a” and “an” refers to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element.

The term “about” when referring to a measurable value such as an amount, a temporal duration, and the like, is meant to encompass variations of ±20% or in some instances ±10%, or in some instances ±5%, or in some instances ±1%, or in some instances ±0.1% from the specified value, as such variations are appropriate to perform the disclosed methods.

The term “acquire” or “acquiring” as the terms are used herein, refer to obtaining possession of a physical entity (e.g., a sample, a polypeptide, a nucleic acid, or a sequence), or a value, e.g., a numerical value, by “directly acquiring” or “indirectly acquiring” the physical entity or value. “Directly acquiring” means performing a process (e.g., performing a synthetic or analytical method) to obtain the physical entity or value. “Indirectly acquiring” refers to receiving the physical entity or value from another party or source (e.g., a third party laboratory that directly acquired the physical entity or value). Directly acquiring a physical entity includes performing a process that includes a physical change in a physical substance, e.g., a starting material. Directly acquiring a value includes performing a process that includes a physical change in a sample or another substance, e.g., performing an analytical process which includes a physical change in a substance, e.g., a sample.

As used herein, the term “T cell receptor beta variable chain” or “TCRβV,” refers to an extracellular region of the T cell receptor beta chain which comprises the antigen recognition domain of the T cell receptor. The term TCRβV includes isoforms, mammalian, e.g., human TCRβV, species homologs of human and analogs comprising at least one common epitope with TCRβV. Human TCRβV comprises a gene family comprising subfamilies including, but not limited to: a TCRβ V6 subfamily, a TCRβ V10 subfamily, a TCRβ V12 subfamily, a TCRβ V5 subfamily, a TCRβ V7 subfamily, a TCRβ V11 subfamily, a TCRβ V14 subfamily, a TCRβ V16 subfamily, a TCRβ V18 subfamily, a TCRβ V9 subfamily, a TCRβ V13 subfamily, a TCRβ V4 subfamily, a TCRβ V3 subfamily, a TCRβ V2 subfamily, a TCRβ V15 subfamily, a TCRβ V30 subfamily, a TCRβ V19 subfamily, a TCRβ V27 subfamily, a TCRβ V28 subfamily, a TCRβ V24 subfamily, a TCRβ V20 subfamily, TCRβ V25 subfamily, a TCRβ V29 subfamily, a TCRβ V1 subfamily, a TCRβ V17 subfamily, a TCRβ V21 subfamily, a TCRβ V23 subfamily, or a TCRβ V26 subfamily, as well as family members of said subfamilies, and variants thereof (e.g., a structural or functional variant thereof). In some embodiments, the TCRβ V6 subfamily comprises: TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01. In some embodiments, TCRβV comprises TCRβ V6-5*01, or a variant thereof, e.g., a variant having 85%, 90%, 95%, 99% or more identity the naturally-occurring sequence. TCRβ V6-5*01 is also known as TRBV65; TCRBV6S5; TCRBV13S1, or TCRβ V13.1. The amino acid sequence of TCRβ V6-5*01, e.g., human TCRβ V6-5*01, is known in that art, e.g., as provided by IMGT ID L36092. In some embodiments, TCRβ V6-5*01 is encoded by the nucleic acid sequence of SEQ ID NO: 43, or a sequence having 85%, 90%, 95%, 99% or more identity thereof. In some embodiments, TCRβ V6-5*01 comprises the amino acid sequence of SEQ ID NO: 44, or a sequence having 85%, 90%, 95%, 99% or more identity thereof.

The term “human-like antibody molecule” as used herein refers to a humanized antibody molecule, human antibody molecule or an antibody molecule having at least 95% identity with a non-murine germline framework region, e.g., FR1, FR2, FR3 and/or FR4. In some embodiments, the human-like antibody molecule comprises a framework region having at least 95% identity to a human germline framework region, e.g., a FR1, FR2, FR3 and/or FR4 of a human germline framework region. In some embodiments, the human-like antibody molecule is a recombinant antibody. In some embodiments, the human-like antibody molecule is a humanized antibody molecule. In some embodiments, the human-like antibody molecule is human antibody molecule. In some embodiments, the human-like antibody molecule is a phage display or a yeast display antibody molecule. In some embodiments, the human-like antibody molecule is a chimeric antibody molecule. In some embodiments, the human-like antibody molecule is a CDR grafted antibody molecule.

The term “cytokine profile” as used herein, refers to the level and/or activity of on one or more cytokines or chemokines, e.g., as described herein. In some embodiments, a cytokine profile comprises the level and/or activity of a naturally occurring cytokine, a fragment or a variant thereof. In an embodiment, a cytokine profile comprises the level and/or activity of one or more cytokines and/or one or more chemokines (e.g., as described herein). In some embodiments, a cytokine profile comprises the level and/or activity of a naturally occurring cytokine, a fragment or a variant thereof. In some embodiments, a cytokine profile comprises the level and/or activity of a naturally occurring chemokine, a fragment or a variant thereof. In an embodiment, a cytokine profile comprises the level and/or activity of one or more of: IL-2 (e.g., full length, a variant, or a fragment thereof); IL-1beta (e.g., full length, a variant, or a fragment thereof); IL-6 (e.g., full length, a variant, or a fragment thereof); TNFα (e.g., full length, a variant, or a fragment thereof); IFNg (e.g., full length, a variant, or a fragment thereof) IL-10 (e.g., full length, a variant, or a fragment thereof); IL-4 (e.g., full length, a variant, or a fragment thereof); TNF alpha (e.g., full length, a variant, or a fragment thereof); IL-12p70 (e.g., full length, a variant, or a fragment thereof); IL-13 (e.g., full length, a variant, or a fragment thereof); IL-8 (e.g., full length, a variant, or a fragment thereof); Eotaxin (e.g., full length, a variant, or a fragment thereof); Eotaxin-3 (e.g., full length, a variant, or a fragment thereof); IL-8 (HA) (e.g., full length, a variant, or a fragment thereof); IP-10 (e.g., full length, a variant, or a fragment thereof); MCP-1 (e.g., full length, a variant, or a fragment thereof); MCP-4 (e.g., full length, a variant, or a fragment thereof); MDC (e.g., full length, a variant, or a fragment thereof); MIP-1a (e.g., full length, a variant, or a fragment thereof); MIP-1b (e.g., full length, a variant, or a fragment thereof); TARC (e.g., full length, a variant, or a fragment thereof); GM-CSF (e.g., full length, a variant, or a fragment thereof); IL-12 23p40 (e.g., full length, a variant, or a fragment thereof); IL-15 (e.g., full length, a variant, or a fragment thereof); IL-16 (e.g., full length, a variant, or a fragment thereof); IL-17a (e.g., full length, a variant, or a fragment thereof); IL-1a (e.g., full length, a variant, or a fragment thereof); IL-5 (e.g., full length, a variant, or a fragment thereof); IL-7 (e.g., full length, a variant, or a fragment thereof); TNF-beta (e.g., full length, a variant, or a fragment thereof); or VEGF (e.g., full length, a variant, or a fragment thereof). In some embodiments, a cytokine profile includes secretion of one or more cytokines or chemokines.

In an embodiment, a cytokine in a cytokine profile can be modulated, e.g., increased or decreased, by an anti-TCRBV antibody molecule described herein. In one embodiment, the cytokine profile includes cytokines associated with a cytokine storm or cytokine release syndrome (CRS), e.g., IL-6, IL-1beta, TNFalpha and IL-10.

The term “variant” refers to a polypeptide that has a substantially identical amino acid sequence to the naturally-occurring sequence, or are encoded by a substantially identical nucleotide sequence. In some embodiments, the variant is a functional variant. In some embodiments, a TCRβV variant can bind to TCRα and form a TCR α:β complex.

The term “functional variant” refers to a polypeptide that has a substantially identical amino acid sequence to the naturally-occurring sequence, or are encoded by a substantially identical nucleotide sequence, and are capable of having one or more activities of the naturally-occurring sequence.

As used herein, a “multifunctional” or a “multispecific” molecule refers to molecule, e.g., a polypeptide, that has two or more functionalities, e.g., two or more binding specificities. In some embodiments, the functionalities can include one or more immune cell engagers, one or more tumor binding molecules, one or more cytokine molecules, one or more stromal modifiers, and other moieties described herein. In some embodiments, the multispecific molecule is a multispecific antibody molecule, e.g., a bispecific antibody molecule. In some embodiments, the multispecific molecule includes an anti-TCRVb antibody molecule as described herein.

In some embodiments, the multifunctional molecule includes an immune cell engager. “An immune cell engager” refers to one or more binding specificities that bind and/or activate an immune cell, e.g., a cell involved in an immune response. In embodiments, the immune cell is chosen from a T cell, an NK cell, a B cell, a dendritic cell, and/or the macrophage cell. The immune cell engager can be an antibody molecule, a receptor molecule (e.g., a full length receptor, receptor fragment, or fusion thereof (e.g., a receptor-Fc fusion)), or a ligand molecule (e.g., a full length ligand, ligand fragment, or fusion thereof (e.g., a ligand-Fc fusion)) that binds to the immune cell antigen (e.g., the T cell, the NK cell antigen, the B cell antigen, the dendritic cell antigen, and/or the macrophage cell antigen). In embodiments, the immune cell engager specifically binds to the target immune cell, e.g., binds preferentially to the target immune cell. For example, when the immune cell engager is an antibody molecule, it binds to an immune cell antigen (e.g., a T cell antigen, an NK cell antigen, a B cell antigen, a dendritic cell antigen, and/or a macrophage cell antigen) with a dissociation constant of less than about 10 nM.

In some embodiments, the multifunctional molecule includes a cytokine molecule. As used herein, a “cytokine molecule” refers to full length, a fragment or a variant of a cytokine; a cytokine further comprising a receptor domain, e.g., a cytokine receptor dimerizing domain; or an agonist of a cytokine receptor, e.g., an antibody molecule (e.g., an agonistic antibody) to a cytokine receptor, that elicits at least one activity of a naturally-occurring cytokine. In some embodiments the cytokine molecule is chosen from interleukin-2 (IL-2), interleukin-7 (IL-7), interleukin-12 (IL-12), interleukin-10 (IL-10), interleukin-15 (IL-15), interleukin-18 (IL-18), interleukin-21 (IL-21), or interferon gamma, or a fragment or variant thereof, or a combination of any of the aforesaid cytokines. The cytokine molecule can be a monomer or a dimer. In embodiments, the cytokine molecule can further include a cytokine receptor dimerizing domain. In other embodiments, the cytokine molecule is an agonist of a cytokine receptor, e.g., an antibody molecule (e.g., an agonistic antibody) to a cytokine receptor chosen from an IL-15Ra or IL-21R.

As used herein, the term “molecule” as used in, e.g., antibody molecule, cytokine molecule, receptor molecule, includes full-length, naturally-occurring molecules, as well as variants, e.g., functional variants (e.g., truncations, fragments, mutated (e.g., substantially similar sequences) or derivatized form thereof), so long as at least one function and/or activity of the unmodified (e.g., naturally-occurring) molecule remains.

In some embodiments, the multifunctional molecule includes a stromal modifying moiety. A “stromal modifying moiety,” as used herein refers to an agent, e.g., a protein (e.g., an enzyme), that is capable of altering, e.g., degrading a component of, the stroma. In embodiments, the component of the stroma is chosen from, e.g., an ECM component, e.g., a glycosaminoglycan, e.g., hyaluronan (also known as hyaluronic acid or HA), chondroitin sulfate, chondroitin, dermatan sulfate, heparin sulfate, heparin, entactin, tenascin, aggrecan and keratin sulfate; or an extracellular protein, e.g., collagen, laminin, elastin, fibrinogen, fibronectin, and vitronectin.

Certain terms are defined below.

As used herein, the articles “a” and “an” refer to one or more than one, e.g., to at least one, of the grammatical object of the article. The use of the words “a” or “an” when used in conjunction with the term “comprising” herein may mean “one,” but it is also consistent with the meaning of “one or more,” “at least one,” and “one or more than one.”

As used herein, “about” and “approximately” generally mean an acceptable degree of error for the quantity measured given the nature or precision of the measurements. Exemplary degrees of error are within 20 percent (%), typically, within 10%, and more typically, within 5% of a given range of values.

“Antibody molecule” as used herein refers to a protein, e.g., an immunoglobulin chain or fragment thereof, comprising at least one immunoglobulin variable domain structure and/or sequence. An antibody molecule encompasses antibodies (e.g., full-length antibodies) and antibody fragments. In an embodiment, an antibody molecule comprises an antigen binding or functional fragment of a full length antibody, or a full length immunoglobulin chain. For example, a full-length antibody is an immunoglobulin (Ig) molecule (e.g., an IgG antibody) that is naturally occurring or formed by normal immunoglobulin gene fragment recombinatorial processes). In embodiments, an antibody molecule refers to an immunologically active, antigen-binding portion of an immunoglobulin molecule, such as an antibody fragment. An antibody fragment, e.g., functional fragment, is a portion of an antibody, e.g., Fab, Fab′, F(ab′)2, F(ab)2, variable fragment (Fv), domain antibody (dAb), or single chain variable fragment (scFv). A functional antibody fragment binds to the same antigen as that recognized by the intact (e.g., full-length) antibody. The terms “antibody fragment” or “functional fragment” also include isolated fragments consisting of the variable regions, such as the “Fv” fragments consisting of the variable regions of the heavy and light chains or recombinant single chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker (“scFv proteins”). In some embodiments, an antibody fragment does not include portions of antibodies without antigen binding activity, such as Fc fragments or single amino acid residues. Exemplary antibody molecules include full length antibodies and antibody fragments, e.g., dAb (domain antibody), single chain, Fab, Fab′, and F(ab′)2 fragments, and single chain variable fragments (scFvs). In some embodiments, the antibody molecule is an antibody mimetic. In some embodiments, the antibody molecule is, or comprises, an antibody-like framework or scaffold, such as, fibronectins, ankyrin repeats (e.g., designed ankyrin repeat proteins (DARPins)), avimers, affibody affinity ligands, anticalins, or affilin molecules.

As used herein, an “immunoglobulin variable domain sequence” refers to an amino acid sequence which can form the structure of an immunoglobulin variable domain. For example, the sequence may include all or part of the amino acid sequence of a naturally-occurring variable domain. For example, the sequence may or may not include one, two, or more N- or C-terminal amino acids, or may include other alterations that are compatible with formation of the protein structure.

In embodiments, an antibody molecule is monospecific, e.g., it comprises binding specificity for a single epitope. In some embodiments, an antibody molecule is multispecific, e.g., it comprises a plurality of immunoglobulin variable domain sequences, where a first immunoglobulin variable domain sequence has binding specificity for a first epitope and a second immunoglobulin variable domain sequence has binding specificity for a second epitope. In some embodiments, an antibody molecule is a bispecific antibody molecule. “Bispecific antibody molecule” as used herein refers to an antibody molecule that has specificity for more than one (e.g., two, three, four, or more) epitope and/or antigen.

“Antigen” (Ag) as used herein refers to a molecule that can provoke an immune response, e.g., involving activation of certain immune cells and/or antibody generation. Any macromolecule, including almost all proteins or peptides, can be an antigen. Antigens can also be derived from genomic recombinant or DNA. For example, any DNA comprising a nucleotide sequence or a partial nucleotide sequence that encodes a protein capable of eliciting an immune response encodes an “antigen.” In embodiments, an antigen does not need to be encoded solely by a full length nucleotide sequence of a gene, nor does an antigen need to be encoded by a gene at all. In embodiments, an antigen can be synthesized or can be derived from a biological sample, e.g., a tissue sample, a tumor sample, a cell, or a fluid with other biological components. As used, herein a “tumor antigen” or interchangeably, a “cancer antigen” includes any molecule present on, or associated with, a cancer, e.g., a cancer cell or a tumor microenvironment that can provoke an immune response. As used, herein an “immune cell antigen” includes any molecule present on, or associated with, an immune cell that can provoke an immune response.

The “antigen-binding site,” or “binding portion” of an antibody molecule refers to the part of an antibody molecule, e.g., an immunoglobulin (Ig) molecule, that participates in antigen binding. In embodiments, the antigen binding site is formed by amino acid residues of the variable (V) regions of the heavy (H) and light (L) chains. Three highly divergent stretches within the variable regions of the heavy and light chains, referred to as hypervariable regions, are disposed between more conserved flanking stretches called “framework regions,” (FRs). FRs are amino acid sequences that are naturally found between, and adjacent to, hypervariable regions in immunoglobulins. In embodiments, in an antibody molecule, the three hypervariable regions of a light chain and the three hypervariable regions of a heavy chain are disposed relative to each other in three dimensional space to form an antigen-binding surface, which is complementary to the three-dimensional surface of a bound antigen. The three hypervariable regions of each of the heavy and light chains are referred to as “complementarity-determining regions,” or “CDRs.” The framework region and CDRs have been defined and described, e.g., in Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242, and Chothia, C. et al. (1987) J. Mol. Biol. 196:901-917. Each variable chain (e.g., variable heavy chain and variable light chain) is typically made up of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the amino acid order: FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4.

“Cancer” as used herein can encompass all types of oncogenic processes and/or cancerous growths. In embodiments, cancer includes primary tumors as well as metastatic tissues or malignantly transformed cells, tissues, or organs. In embodiments, cancer encompasses all histopathologies and stages, e.g., stages of invasiveness/severity, of a cancer. In embodiments, cancer includes relapsed and/or resistant cancer. The terms “cancer” and “tumor” can be used interchangeably. For example, both terms encompass solid and liquid tumors. As used herein, the term “cancer” or “tumor” includes premalignant, as well as malignant cancers and tumors.

As used herein, an “immune cell” refers to any of various cells that function in the immune system, e.g., to protect against agents of infection and foreign matter. In embodiments, this term includes leukocytes, e.g., neutrophils, eosinophils, basophils, lymphocytes, and monocytes. Innate leukocytes include phagocytes (e.g., macrophages, neutrophils, and dendritic cells), mast cells, eosinophils, basophils, and natural killer cells. Innate leukocytes identify and eliminate pathogens, either by attacking larger pathogens through contact or by engulfing and then killing microorganisms, and are mediators in the activation of an adaptive immune response. The cells of the adaptive immune system are special types of leukocytes, called lymphocytes. B cells and T cells are important types of lymphocytes and are derived from hematopoietic stem cells in the bone marrow. B cells are involved in the humoral immune response, whereas T cells are involved in cell-mediated immune response. The term “immune cell” includes immune effector cells.

“Immune effector cell,” as that term is used herein, refers to a cell that is involved in an immune response, e.g., in the promotion of an immune effector response. Examples of immune effector cells include, but are not limited to, T cells, e.g., alpha/beta T cells and gamma/delta T cells, B cells, natural killer (NK) cells, natural killer T (NK T) cells, and mast cells.

The term “effector function” or “effector response” refers to a specialized function of a cell. Effector function of a T cell, for example, may be cytolytic activity or helper activity including the secretion of cytokines.

The compositions and methods of the present invention encompass polypeptides and nucleic acids having the sequences specified, or sequences substantially identical or similar thereto, e.g., sequences at least 80%, 85%, 90%, 95% identical or higher to the sequence specified. In the context of an amino acid sequence, the term “substantially identical” is used herein to refer to a first amino acid that contains a sufficient or minimum number of amino acid residues that are i) identical to, or ii) conservative substitutions of aligned amino acid residues in a second amino acid sequence such that the first and second amino acid sequences can have a common structural domain and/or common functional activity. For example, amino acid sequences that contain a common structural domain having at least about 80%, 85%, 90%. 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity to a reference sequence, e.g., a sequence provided herein.

In the context of nucleotide sequence, the term “substantially identical” is used herein to refer to a first nucleic acid sequence that contains a sufficient or minimum number of nucleotides that are identical to aligned nucleotides in a second nucleic acid sequence such that the first and second nucleotide sequences encode a polypeptide having common functional activity, or encode a common structural polypeptide domain or a common functional polypeptide activity. For example, nucleotide sequences having at least about 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity to a reference sequence, e.g., a sequence provided herein.

The term “variant” refers to a polypeptide that has a substantially identical amino acid sequence to a reference amino acid sequence, or is encoded by a substantially identical nucleotide sequence. In some embodiments, the variant is a functional variant.

The term “functional variant” refers to a polypeptide that has a substantially identical amino acid sequence to a reference amino acid sequence, or is encoded by a substantially identical nucleotide sequence, and is capable of having one or more activities of the reference amino acid sequence.

Calculations of homology or sequence identity between sequences (the terms are used interchangeably herein) are performed as follows.

To determine the percent identity of two amino acid sequences, or of two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes). In a preferred embodiment, the length of a reference sequence aligned for comparison purposes is at least 30%, preferably at least 40%, more preferably at least 50%, 60%, and even more preferably at least 70%, 80%, 90%, 100% of the length of the reference sequence. The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position (as used herein amino acid or nucleic acid “identity” is equivalent to amino acid or nucleic acid “homology”).

The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.

The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. In a preferred embodiment, the percent identity between two amino acid sequences is determined using the Needleman and Wunsch ((1970) J Mol. Biol. 48:444-453) algorithm which has been incorporated into the GAP program in the GCG software package (available at http://www.gcg.com), using either a Blossum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6. In yet another preferred embodiment, the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package (available at http://www.gcg.com), using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6. A particularly preferred set of parameters (and the one that should be used unless otherwise specified) are a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5.

The percent identity between two amino acid or nucleotide sequences can be determined using the algorithm of E. Meyers and W. Miller ((1989) CABIOS, 4:11-17) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.

The nucleic acid and protein sequences described herein can be used as a “query sequence” to perform a search against public databases to, for example, identify other family members or related sequences. Such searches can be performed using the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. (1990) J. Mol. Biol. 215:403-10. BLAST nucleotide searches can be performed with the NBLAST program, score=100, wordlength=12 to obtain nucleotide sequences homologous to a nucleic acid molecule of the invention. BLAST protein searches can be performed with the XBLAST program, score=50, wordlength=3 to obtain amino acid sequences homologous to protein molecules of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al., (1997) Nucleic Acids Res. 25:3389-3402. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used.

It is understood that the molecules of the present invention may have additional conservative or non-essential amino acid substitutions, which do not have a substantial effect on their functions.

The term “amino acid” is intended to embrace all molecules, whether natural or synthetic, which include both an amino functionality and an acid functionality and capable of being included in a polymer of naturally-occurring amino acids. Exemplary amino acids include naturally-occurring amino acids; analogs, derivatives and congeners thereof; amino acid analogs having variant side chains; and all stereoisomers of any of any of the foregoing. As used herein the term “amino acid” includes both the D- or L-optical isomers and peptidomimetics.

A “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).

The terms “polypeptide”, “peptide” and “protein” (if single chain) are used interchangeably herein to refer to polymers of amino acids of any length. The polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids. The terms also encompass an amino acid polymer that has been modified; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation, such as conjugation with a labeling component. The polypeptide can be isolated from natural sources, can be a produced by recombinant techniques from a eukaryotic or prokaryotic host, or can be a product of synthetic procedures.

The terms “nucleic acid,” “nucleic acid sequence,” “nucleotide sequence,” or “polynucleotide sequence,” and “polynucleotide” are used interchangeably. They refer to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or analogs thereof. The polynucleotide may be either single-stranded or double-stranded, and if single-stranded may be the coding strand or non-coding (antisense) strand. A polynucleotide may comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs. The sequence of nucleotides may be interrupted by non-nucleotide components. A polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component. The nucleic acid may be a recombinant polynucleotide, or a polynucleotide of genomic, cDNA, semisynthetic, or synthetic origin which either does not occur in nature or is linked to another polynucleotide in a non-natural arrangement.

The term “isolated,” as used herein, refers to material that is removed from its original or native environment (e.g., the natural environment if it is naturally occurring). For example, a naturally-occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or polypeptide, separated by human intervention from some or all of the co-existing materials in the natural system, is isolated. Such polynucleotides could be part of a vector and/or such polynucleotides or polypeptides could be part of a composition, and still be isolated in that such vector or composition is not part of the environment in which it is found in nature.

Various aspects of the invention are described in further detail below. Additional definitions are set out throughout the specification.

Human T Cell Receptor (TCR) Complex

T cell receptors (TCR) can be found on the surface of T cells. TCRs recognize antigens, e.g., peptides, presented on, e.g., bound to, major histocompatibility complex (MHC) molecules on the surface of cells, e.g., antigen-presenting cells. TCRs are heterodimeric molecules and can comprise an alpha chain, a beta chain, a gamma chain or a delta chain. TCRs comprising an alpha chain and a beta chain are also referred to as TCRαβ. The TCR beta chain consists of the following regions (also known as segments): variable (V), diversity (D), joining (J) and constant (C) (see Mayer G. and Nyland J. (2010) Chapter 10: Major Histocompatibility Complex and T-cell Receptors-Role in Immune Responses. In: Microbiology and Immunology on-line, University of South Carolina School of Medicine). The TCR alpha chain consists of V, J and C regions. The rearrangement of the T-cell receptor (TCR) through somatic recombination of V (variable), D (diversity), J (joining), and C (constant) regions is a defining event in the development and maturation of a T cell. TCR gene rearrangement takes place in the thymus.

TCRs can comprise a receptor complex, known as the TCR complex, which comprises a TCR heterodimer comprising of an alpha chain and a beta chain, and dimeric signaling molecules, e.g., CD3 co-receptors, e.g., CD3δ/ε, and/or CD3γ/ε.

TCR Beta V (TCRβV)

Diversity in the immune system enables protection against a huge array of pathogens. Since the germline genome is limited in size, diversity is achieved not only by the process of V(D)J recombination but also by junctional (junctions between V-D and D-J segments) deletion of nucleotides and addition of pseudo-random, non-templated nucleotides. The TCR beta gene undergoes gene arrangement to generate diversity.

The TCR V beta repertoire varies between individuals and populations because of, e.g., 7 frequently occurring inactivating polymorphisms in functional gene segments and a large insertion/deletion-related polymorphism encompassing 2 V beta gene segments.

This disclosure provides, inter alia, antibody molecules and fragments thereof, that bind, e.g., specifically bind, to a human TCR beta V chain (TCRβV), e.g., a TCRβV gene family (also referred to as a group), e.g., a TCRβV subfamily (also referred to as a subgroup), e.g., as described herein. TCR beta V families and subfamilies are known in the art, e.g., as described in Yassai et al., (2009) Immunogenetics 61(7)pp:493-502; Wei S. and Concannon P. (1994) Human Immunology 41(3) pp: 201-206. The antibodies described herein can be recombinant antibodies, e.g., recombinant non-murine antibodies, e.g., recombinant human or humanized antibodies.

The terms TCRBV, TCRVB, TRBV, TCRβV, TCRVβ or TRW are used interchangeably herein and refer to a TCR beta V chain, e.g., as described herein.

In an aspect, the disclosure provides an anti-TCRβV antibody molecule that binds to human TCRβV, e.g., a TCRβV family, e.g., gene family or a variant thereof. In some embodiments a TCRBV gene family comprises one or more subfamilies, e.g., as described herein, e.g., in FIG. 3, Table 8A or Table 8B. In some embodiments, the TCRβV gene family comprises: a TCRβ V6 subfamily, a TCRβ V10 subfamily, a TCRβ V12 subfamily, a TCRβ V5 subfamily, a TCRβ V7 subfamily, a TCRβ V11 subfamily, a TCRβ V14 subfamily, a TCRβ V16 subfamily, a TCRβ V18 subfamily, a TCRβ V9 subfamily, a TCRβ V13 subfamily, a TCRβ V4 subfamily, a TCRβ V3 subfamily, a TCRβ V2 subfamily, a TCRβ V15 subfamily, a TCRβ V30 subfamily, a TCRβ V19 subfamily, a TCRβ V27 subfamily, a TCRβ V28 subfamily, a TCRβ V24 subfamily, a TCRβ V20 subfamily, TCRβ V25 subfamily, a TCRβ V29 subfamily, a TCRβ V1 subfamily, a TCRβ V17 subfamily, a TCRβ V21 subfamily, a TCRβ V23 subfamily, or a TCRβ V26 subfamily.

In some embodiments, TCRβ V6 subfamily is also known as TCRβ V13.1. In some embodiments, the TCRβ V6 subfamily comprises: TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-4*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-4*02, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-9*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-8*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-5*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-6*02, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-6*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-2*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-3 *01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-1*01, or a variant thereof.

In some embodiments, TCRβ V6 comprises TCRβ V6-5*01, or a variant thereof. In some embodiments, TCRβ V6, e.g., TCRβ V6-5*01, is recognized, e.g., bound, by SEQ ID NO: 1 and/or SEQ ID NO: 2. In some embodiments, TCRβ V6, e.g., TCRβ V6-5*01, is recognized, e.g., bound, by SEQ ID NO: 9 and/or SEQ ID NO: 10. In some embodiments, TCRβ V6 is recognized, e.g., bound, by SEQ ID NO: 9 and/or SEQ ID NO: 11.

In some embodiments, TCRβ V10 subfamily is also known as TCRβ V12. In some embodiments, the TCRβ V10 subfamily comprises: TCRβ V10-1*01, TCRβ V10-1*02, TCRβ V10-3*01 or TCRβ V10-2*01, or a variant thereof.

In some embodiments, TCRβ V12 subfamily is also known as TCRβ V8.1. In some embodiments, the TCRβ V12 subfamily comprises: TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01, or a variant thereof. In some embodiments, TCRβ V12 is recognized, e.g., bound, by SEQ ID NO: 15 and/or SEQ ID NO: 16. In some embodiments, TCRβ V12 is recognized, e.g., bound, by any one of SEQ ID NOs 23-25, and/or any one of SEQ ID NO: 26-30:

In some embodiments, the TCRβ V5 subfamily is chosen from: TCRβ V5-5*01, TCRβ V5-6*01, TCRβ V5-4*01, TCRβ V5-8*01, TCRβ V5-1*01, or a variant thereof.

In some embodiments, the TCRβ V7 subfamily comprises TCRβ V7-7*01, TCRβ V7-6*01, TCRβ V7-8*02, TCRβ V7-4*01, TCRβ V7-2*02, TCRβ V7-2*03, TCRβ V7-2*01, TCRβ V7-3 *01, TCRβ V7-9*03, or TCRβ V7-9*01, or a variant thereof.

In some embodiments, the TCRβ V11 subfamily comprises: TCRβ V11-1*01, TCRβ V11-2*01 or TCRβ V11-3*01, or a variant thereof.

In some embodiments, the TCRβ V14 subfamily comprises TCRβ V14*01, or a variant thereof.

In some embodiments, the TCRβ V16 subfamily comprises TCRβ V16*01, or a variant thereof.

In some embodiments, the TCRβ V18 subfamily comprises TCRβ V18*01, or a variant thereof.

In some embodiments, the TCRβ V9 subfamily comprises TCRβ V9*01 or TCRβ V9*02, or a variant thereof.

In some embodiments, the TCRβ V13 subfamily comprises TCRβ V13*01, or a variant thereof.

In some embodiments, the TCRβ V4 subfamily comprises TCRβ V4-2*01, TCRβ V4-3*01, or TCRβ V4-1*01, or a variant thereof.

In some embodiments, the TCRβ V3 subfamily comprises TCRβ V3-1*01, or a variant thereof.

In some embodiments, the TCRβ V2 subfamily comprises TCRβ V2*01, or a variant thereof.

In some embodiments, the TCRβ V15 subfamily comprises TCRβ V15*01, or a variant thereof.

In some embodiments, the TCRβ V30 subfamily comprises TCRβ V30*01, or TCRβ V30*02, or a variant thereof.

In some embodiments, the TCRβ V19 subfamily comprises TCRβ V19*01, or TCRβ V19*02, or a variant thereof.

In some embodiments, the TCRβ V27 subfamily comprises TCRβ V27*01, or a variant thereof.

In some embodiments, the TCRβ V28 subfamily comprises TCRβ V28*01, or a variant thereof.

In some embodiments, the TCRβ V24 subfamily comprises TCRβ V24-1*01, or a variant thereof.

In some embodiments, the TCRβ V20 subfamily comprises TCRβ V20-1*01, or TCRβ V20-1*02, or a variant thereof.

In some embodiments, the TCRβ V25 subfamily comprises TCRβ V25-1*01, or a variant thereof.

In some embodiments, the TCRβ V29 subfamily comprises TCRβ V29-1*01, or a variant thereof.

TABLE 8A List of TCRβV subfamilies and subfamily members Reference in FIG. 3 Subfamily Subfamily members A TCRβ V6 TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, Also referred to as: TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCR VB 13.1 TCRβ V6-3*01 or TCRβ V6-1*01. B TCRβ V10 TCRβ V10-1*01, TCRβ V10-1*02, TCRβ V10-3*01 or TCRβ V10- Also referred to as: 2*01 TCRβ V12 C TCRβ V12 TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01 Also referred to as: TCRβ V8.1 D TCRβ V5 TCRβ V5-5*01, TCRβ V5-6*01, TCRβ V5-4*01, TCRβ V5-8*01, TCRβ V5-1*01 E TCRβ V7 TCRβ V7-7*01, TCRβ V7-6*01, TCRβ V7 -8*02, TCRβ V7 -4*01, TCRβ V7-2*02, TCRβ V7-2*03, TCRβ V7-2*01, TCRβ V7-3*01, TCRβ V7-9*03, or TCRβ V7-9*01 F TCRβ V11 TCRβ V11-1*01, TCRβ V11-2*01 or TCRβ V11-3*01 G TCRβ V14 TCRβ V14*01 H TCRβ V16 TCRβ V16*01 I TCRβ V18 TCRβ V18*01 J TCRβ V9 TCRβ V9*01 or TCRβ V9*02 K TCRβ V13 TCRβ V13*01 L TCRβ V4 TCRβ V4-2*01, TCRβ V4-3*01, or TCRβ V4-1*01 M TCRβ V3 TCRβ V3-1*01 N TCRβ V2 TCRβ V2*01 O TCRβ V15 TCRβ V15*01 P TCRβ V30 TCRβ V30*01, or TCRβ V30*02 Q TCRβ V19 TCRβ V19*01, or TCRβ V19*02 R TCRβ V27 TCRβ V27*01. S TCRβ V28 TCRβ V28*01. T TCRβ V24 TCRβ V24-1*01 U TCRβ V20 TCRβ V20-1*01, or TCRβ V20-1*02 V TCRβ V25 TCRβ V25-1*01 W TCRβ V29 TCRβ V29-1*01

TABLE 8B Additional TCRβV subfamilies Subfamily TCRβ V1 TCRβ V17 TCRβ V21 TCRβ V23 TCRβ V26

The various TCRβV subfamilies and/or subfamily members can be expressed at different levels in individuals, e.g., healthy individuals, as disclosed in Kitaura K. et al (2016), BMC Immunology vol 17: 38, the entire contents of which are hereby incorporated by reference. For example, TCRβ V6-5 is represented in approximately 3-6% healthy donors.

The representation of various TCRBV subfamilies and/or subfamily members can also be different in cancer cells. For example, TCRβV is present in about 3-6% of tumor infiltrating T cells irrespective of tumor type (see Li B. et al., Nature Genetics, 2016, vol:48(7):725-32 the entire contents of which are hereby incorporated by references). Li et al., also disclose that TCRβ V6-5 is present at a high frequency in tumor cells.

Exemplary amino acid sequences for TCRβV subfamily members can be found on the ImMunoGeneTics Information System website: www.imgt.org, or in a similar resource.

The alignment of TCRBV amino acid sequences in Table 9 underscores the diversity of TCR sequences. In particular, the TRBV sequences from different subfamilies are considerably different from each other.

Anti-TCRβV Antibodies

Disclosed herein, is the discovery of a novel class of antibodies, i.e. anti-TCRβV antibody molecules disclosed herein, which despite having low sequence similarity (e.g., low sequence identity among the different antibody molecules that recognize different TCRβV subfamilies), recognize a structurally conserved region, e.g., domain, on the TCRβV protein (e.g., as denoted by the circled area in FIG. 24A) and have a similar function (e.g., a similar cytokine profile). Thus, the anti-TCRβV antibody molecules disclosed herein share a structure-function relationship.

Without wishing to be bound by theory, it is believed that in some embodiments, the anti-TCRβV antibody molecules disclosed herein bind to an outward facing epitope of a TCRβV protein when it is in a complex with a TCRalpha protein, e.g., as described by the circled area in FIG. 24A. In some embodiments, the anti-TCRβV antibody molecules disclosed herein recognize (e.g., bind to), a structurally conserved domain on the TCRβV protein (e.g., as denoted by the circled area in FIG. 24A).

In some embodiments, the anti-TCRβV antibody molecules disclosed herein do not recognize, e.g., bind to, an interface of a TCRβV:TCRalpha complex.

In some embodiments, the anti-TCRβV antibody molecules disclosed herein do not recognize, e.g., bind to, a constant region of a TCRβV protein. An exemplary antibody that binds to a constant region of a TCRBV region is JOVI.1 as described in Viney et al., (Hybridoma. 1992 December; 11(6):701-13).

In some embodiments, the anti-TCRβV antibody molecules disclosed herein do not recognize, e.g., bind to, one or more (e.g., all) of a complementarity determining region (e.g., CDR1, CDR2 and/or CDR3) of a TCRβV protein.

In some embodiments, the anti-TCRβV antibody molecules disclosed herein binds (e.g., specifically binds) to a TCRβV region. In some embodiments, binding of anti-TCRβV antibody molecules disclosed herein results in a cytokine profile that differs from a cytokine profile of a T cell engager that binds to a receptor or molecule other than a TCRβV region (“a non-TCRβV-binding T cell engager”). In some embodiments, the non-TCRβV-binding T cell engager comprises an antibody that binds to a CD3 molecule (e.g., CD3 epsilon (CD3e) molecule); or a TCR alpha (TCRα) molecule. In some embodiments, the non-TCRβV-binding T cell engager is an OKT3 antibody or an SP34-2 antibody.

In an aspect, the disclosure provides an anti-TCRβV antibody molecule that binds to human TCRβV, e.g., a TCRβV gene family, e.g., one or more of a TCRβV subfamily, e.g., as described herein, e.g., in FIG. 3, Table 8A, or Table 8B. In some embodiments, the anti-TCRβV antibody molecule binds to one or more TCRβV subfamilies chosen from: a TCRβ V6 subfamily, a TCRβ V10 subfamily, a TCRβ V12 subfamily, a TCRβ V5 subfamily, a TCRβ V7 subfamily, a TCRβ V11 subfamily, a TCRβ V14 subfamily, a TCRβ V16 subfamily, a TCRβ V18 subfamily, a TCRβ V9 subfamily, a TCRβ V13 subfamily, a TCRβ V4 subfamily, a TCRβ V3 subfamily, a TCRβ V2 subfamily, a TCRβ V15 subfamily, a TCRβ V30 subfamily, a TCRβ V19 subfamily, a TCRβ V27 subfamily, a TCRβ V28 subfamily, a TCRβ V24 subfamily, a TCRβ V20 subfamily, TCRβ V25 subfamily, a TCRβ V29 subfamily, a TCRβ V1 subfamily, a TCRβ V17 subfamily, a TCRβ V21 subfamily, a TCRβ V23 subfamily, or a TCRβ V26 subfamily, or a variant thereof.

In some embodiments, the anti-TCRβV antibody molecule binds to a TCRβ V6 subfamily comprising: TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01, or a variant thereof. In some embodiments the TCRβ V6 subfamily comprises TCRβ V6-5*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-4*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-4*02, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-9*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-8*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-5*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-6*02, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-6*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-2*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-3 *01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-1*01, or a variant thereof.

In some embodiments, the anti-TCRβV antibody molecule binds to a TCRβ V10 subfamily comprising: TCRβ V10-1*01, TCRβ V10-1*02, TCRβ V10-3*01 or TCRβ V10-2*01, or a variant thereof.

In some embodiments, the anti-TCRβV antibody molecule binds to a TCRβ V12 subfamily comprising: TCRβ V12-4*01, TCRβ V12-3*01 or TCRβ V12-5*01, or a variant thereof.

In some embodiments, the anti-TCRβV antibody molecule binds to a TCRβ V5 subfamily comprising: TCRβ V5-5*01, TCRβ V5-6*01, TCRβ V5-4*01, TCRβ V5-8*01, TCRβ V5-1*01, or a variant thereof.

Exemplary anti-TCRβV antibody molecules and the corresponding TCRβV subfamily recognized by said anti-TCRβV antibody molecules is disclosed in Table 10A.

TABLE 10A Exemplary anti-TCRβV antibody molecules TRBV TRBV Reagents monoclonal antibodies gene name allele name Clone name and Specificity Company product Isotype TRBV2 TRBV2*01 IsMMU 546 (TRBV2) Serotec V BETA 22 Mouse TRBV2*02 Coulter Vbeta22 IgG1 TRBV2*03 TRBV3-1 TRBV3-1*01 FIN9 (TRBV3-1) Serotec Vbeta9 Mouse AMKB1-2 (TRBV3-1) Coulter Vbeta9 IgG2a TRBV3-1*02 BD Biosciences Vbeta9 Mouse IgG1 TRBV4-1 TRBV4-1*01 ZOE (TRBV4-1, TRBV4-2, Serotec V BETA 7 Mouse TRBV4-3) Coulter Vbeta7 IgG2a TRBV4-1*02 3G5 (TRBV4-1) Pierce EndogenV beta 7.1 Mouse IgG2b TRBV4-2 TRBV4-2*01 ZOE (TRBV4-1, TRBV4-2, Serotec V BETA 7 Mouse TRBV4-2*02 TRBV4-3) Coulter Vbeta7 IgG2a TRBV4-3 TRBV4-3*01 ZOE (TRBV4-1, TRBV4-2, Serotec V BETA 7 Mouse TRBV4-3*02 TRBV4-3) Coulter Vbeta7 IgG2a TRBV4-3*03 TRBV4-3*04 ZIZOU4 (TRBV4-3) Coulter Vbeta7.2 Mouse IgG2a TRBV5-1 TRBV5-1*01 IMMU157 (TRBV5-1) Serotec Vbeta5.1 Mouse Coulter Vbeta5.1 IgG2a TRBV5-1*02 LC4 (TRBV5-1) Pierce Endogen V beta 5(c) Mouse BD Biosciences IgGl Vbeta5(c) TRBV5-4 TRBV5-4*01 TRBV5-4*02 TRBV5-4*03 TRBV5-4*04 TRBV5-5 TRBV5-5*01 3D11 (TRBV5-5) Serotec VBETA5.3 Mouse 1C1 (TRBV5-5, TRBV5-6) Coulter Vbeta5.3 IgG1 TRBV5-5*02 W112 (TRBV5-5) Pierce Endogen V beta 5(a) Mouse MH3-2 (TRBV5-5, TRBV5-6) BD Biosciences IgG1 Vbeta5(a) TRBV5-5*03 Pierce Endogen V beta 5(b) Mouse Serotec V beta 5.2/5.3 IgG1 BD Biosciences Vbeta5(b) BD Biosciences Vbeta5 Mouse IgG2a 4H11 (TM27) as disclosed in U.S. Pat. No. 5,861,155 TRBV5-6 TRBV5-6*01 36213 (TRBV5-6) Serotec Vbeta 5.2 Mouse 1C1 (TRBV5-5, TRBV5-6) BD Bioscience IgG1 MH3-2 (TRBV5-5, TRBV5-6) Vbeta5(a) Mouse BD Biosciences Vbeta5 IgG1 Mouse IgG2a TRBV5-8 TRBV5-8*01 TRBV5-8*02 TRBV6-1 TRBV6-1*01 BAM13 (TRBV6-1, TRBV6-5) Pierce Endogen V beta 13 Mouse BD Biosciences IgG1 Vbeta13.1, 13.3 TRBV6-2 TRBV6-2*01 H132 Coulter Vbeta13.2 Mouse IgG1 TRBV6-3 TRBV6-3*01 TRBV6-4 TRBV6-4*01 TRBV6-4*02 TRBV6-5 TRBV6-5*01 IMMU 222 (TRBV6-5, Serotec V BETA 13.1 Mouse TRBV6-6 and TRBV6-9) Coulter Vbeta13.1 IgG2b BAM13 (TRBV6-1, TRBV6-5) Pierce Endogen V beta 13 Mouse BD Biosciences IgG1 Vbeta 13.1, 13.3 TRBV6-6 TRBV6-6*01 JU-74 (TRBV6-6) Serotec Vbeta 13.6 Mouse TRBV6-6*02 JU74.3 (TRBV6-6) Coulter Vbeta 13.6 IgG1 TRBV6-6*03 TRBV6-6*04 IMMU 222 (TRBV6-5, Serotec V BETA 13.1 Mouse TRBV6-6*05 TRBV6-6 and TRBV6-9) Coulter Vbeta13.1 IgG2b TRBV6-8 TRBV6-8*01 TRBV6-9 TRBV6-9*01 IMMU 222 (TRBV6-5, Serotec VBETA 13.1 Mouse TRBV6-6 and TRBV6-9) Coulter Vbeta 13.1 IgG2b TRBV7-2 TRBV7-2*01 OT145 (TRBV7-2) Pierce Endogen V beta 6.7 Mouse TRBV7-2*02 BD Biosciences Vbeta6.7 IgG1 TRBV7-2*03 TRBV7-2*04 TRBV7-3 TRBV7-3*01 TRBV7-3*04 TRBV7-3*05 TRBV7-4 TRBV7-4*01 TRBV7-6 TRBV7-6*01 TRBV7-6*02 TRBV7-7 TRBV7-7*01 TRBV7-7*02 TRBV7-8 TRBV7-8*01 TRBV7-8*02 TRBV7-8*03 TRBV7-9 TRBV7-9*01 TRBV7-9*02 TRBV7-9*03 TRVB7-9*04 TRBV7-9*05 TRBV7-9*06 TRBV7-9*07 TRBV9 TRBV9*01 BL37.2 (TRBV9) Serotec Vbeta1 Rat TRBV9*02 Coulter Vbeta1 IgG1 TRBV9*03 TRBV10-1 TRBV10-1*01 S511 (TRBV10-1, TRBV10-2, Pierce Endogen V beta 12 Mouse TRBV10-1*02 TRBV10-3) BD Biosciences Vbeta12 IgG2b TRBV10-2 TRBV10-2*01 TRBV10-2*02 TRBV10-3 TRBV10-3*01 VER2.32.1 (TRBV10-3) Serotec Vbeta12 Mouse TRBV10-3*02 S511 (TRBV10-1, TRBV10-2, Coulter Vbeta12 IgG2a TRBV10-3*03 TRBV10-3) Pierce Endogen V beta 12 Mouse TRBV10-3*04 BD Biosciences Vbeta12 IgG2b TRBV11-1 TRBV11-1*01 TRBV11-2 TRBV11-2*01 IG125 (TRBV 11-2) Serotec Vbeta21.3 Mouse TRBV11-2*02 Coulter Vbeta21.3 IgG2a TRBV11-2*03 TRBV11-3 TRBV11-3*01 TRBV11-3*02 TRBV11-3*03 TRBV11-3*04 TRBV12-3 TRBV12-3*01 56C5 (TRBV12-3, TRBV12-4) Serotec Vbeta8.1/8.2 Mouse 56C5.2 (TRBV 12-3, TRBV 12-4) Coulter Vbeta8 IgG2a TRBV12-4 TRBV12-4*01 16G8 (TRBV12-3, TRBV12-4) Pierce Endogen V beta 8(a) Mouse MX-6 (TRBV12-3, TRBV12-4) BD Biosciences Vbeta8 IgG2b TRBV12-4*02 JR2 (TRBV 12-3, TRBV 12-4, Pierce Endogen V beta 8(b) Mouse TRBV12-5) IgG2a BD Biosciences Vbeta8 Mouse IgG2b TRBV12-5 TRBV12-5*01 JR2 (TRBV 12-3, TRBV 12-4, BD Biosciences Vbeta8 Mouse TRBV12-5) IgG2b TRBV13 TRBV13*01 AF-23 (TRBV13) Serotec Vbeta23 Mouse TRBV13*02 AF23 (TRBV13) Coulter Vbeta23 AHUT7 (Vbeta23) BD Biosciences Vbeta23 IgG1 TRBV14 TRBV14*01 TAMAYA1.2 (TRBV14) Serotec Vbeta16 Mouse TRBV14*02 Coulter Vbeta16 IgG1 TRBV15 TRBV15*01 TRBV15*02 TRBV15*03 TRBV16 TRBV16*01 TRBV16*03 TRBV18 TRBV18*01 BA62 (TRBV 18) Serotec V BETA 18 Mouse BA62.6 (TRBV 18) Coulter Vbeta18 IgG1 TRBV19 TRBV19*01 C1 (TRBV 19) Pierce Endogen V beta 17 Mouse E17.5F3 (TRBV19) BD Biosciences Vbeta17 IgG1 TRBV19*02 E17.5F3.15.13 (TRBV19) Serotec Vbeta17 Mouse TRBV19*03 Coulter Vbeta17 IgG1 TRBV20-1 TRBV20-1*01 MPB2D5 (TRBV20-1) Serotec VBETA2 Mouse TRBV20-1*02 Coulter Vbeta2 IgG1 TRBV20-1*03 TRBV20-1*04 TRBV20-1*05 TRBV20-1*06 TRBV20-1*07 TRBV24-1 TRBV24-1*01 TRBV25-1 TRBV25-1*01 C21 (TRBV25-1) Serotec V BETA 11 Mouse Coulter Vbeta11 IgG2a TRBV27 TRBV27*01 CAS1.1.3 (TRBV27) Serotec Vbeta14 Mouse Coulter Vbeta14 IgG1 TRBV28 TRBV28*01 CH92 (TRBV28) Serotec Vbeta3 Mouse 8F10 (TRBV28) Coulter Vbeta3 IgM JOVI-3 (TRBV28) Pierce Endogen V beta 3.1 Mouse IgG1 BD Biosciences Vbeta3 Mouse IgG2a TRBV29-1 TRBV29-1*01 WJF24 Coulter Vbeta4 Rat TRBV29-1*02 IgM TRBV29-1*03 TRBV30 TRBV30*01 ELL1.4 (TRBV30) Serotec Vbeta20 Mouse TRBV30*02 Coulter Vbeta20 IgG1 TRBV30*04 TRBV30*05

In some embodiments, the anti-TCRβV antibody molecule does not bind to TCRβ V12, or binds to TCRβ V12 with an affinity and/or binding specificity that is less than (e.g., less than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the 16G8 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments, the anti-TCRβV antibody molecule binds to TCRβ V12 with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the 16G8 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments, the anti-TCRβV antibody molecule binds to a TCRβV region other than TCRβ V12 (e.g., TCRβV region as described herein, e.g., TCRβ V6 subfamily (e.g., TCRβ V6-5*01) with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the 16G8 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments, the anti-TCRβV antibody molecule does not bind to TCRβ V5-5*01 or TCRβ V5-1*01, or binds to TCRβ V5-5*01 or TCRβ V5-1*01 with an affinity and/or binding specificity that is less than (e.g., less than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the TM23 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments, the anti-TCRβV antibody molecule binds to TCRβ V5-5*01 or TCRβ V5-1*00 with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the TM23 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments, the anti-TCRβV antibody molecule binds to a TCRβV region other than TCRβ V5-5*01 or TCRβ V5-1*01 (e.g., TCRβV region as described herein, e.g., TCRβ V6 subfamily (e.g., TCRβ V6-5*01) with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the TM23 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

Anti-TCRβ V6 Antibodies

Accordingly, in one aspect, the disclosure provides an anti-TCRβV antibody molecule that binds to human TCRβ V6, e.g., a TCRβ V6 subfamily comprising: TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01. In some embodiments the TCRβ V6 subfamily comprises TCRβ V6-5*01 or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-4*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-4*02, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-9*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-8*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-5*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-6*02, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-6*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-2*01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-3 *01, or a variant thereof. In some embodiments, TCRβ V6 comprises TCRβ V6-1*01, or a variant thereof.

In some embodiments, TCRβ V6-5*01 is encoded by the nucleic acid sequence of SEQ ID NO: 43, or a sequence having 85%, 90%, 95%, 99% or more identity thereof.

SEQ ID NO: 43 ATGAGCATCGGCCTCCTGTGCTGTGCAGCCTTGTCTCTCCTGTGGGCAG GTCCAGTGAATGCTGGTGTCACTCAGACCCCAAAATTCCAGGTCCTGAA GACAGGACAGAGCATGACACTGCAGTGTGCCCAGGATATGAACCATGAA TACATGTCCTGGTATCGACAAGACCCAGGCATGGGGCTGAGGCTGATTC ATTACTCAGTTGGTGCTGGTATCACTGACCAAGGAGAAGTCCCCAATGG CTACAATGTCTCCAGATCAACCACAGAGGATTTCCCGCTCAGGCTGCTG TCGGCTGCTCCCTCCCAGACATCTGTGTACTTCTGTGCCAGCAGTTACT C

In some embodiments, TCRβ V6-5*01 comprises the amino acid sequence of SEQ ID NO: 44, or an amino acid sequence having 85%, 90%, 95%, 99% or more identity thereof.

SEQ ID NO: 44 MSIGLLCCAALSLLWAGPVNAGVTQTPKFQVLKTGQSMTLQCAQDMNHE YMSWYRQDPGMGLRLIHYSVGAGITDQGEVPNGYNVSRSTTEDFPLRLL SAAPSQTSVYFCASSY

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, is a non-murine antibody molecule, e.g., a human or humanized antibody molecule. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule is a human antibody molecule. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule is a humanized antibody molecule.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, is isolated or recombinant.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises at least one antigen-binding region, e.g., a variable region or an antigen-binding fragment thereof, from an antibody described herein, e.g., an antibody chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, or an antibody described in Table 1, or encoded by a nucleotide sequence in Table 1, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises at least one, two, three or four variable regions from an antibody described herein, e.g., an antibody chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, or an antibody described in Table 1, or encoded by a nucleotide sequence in Table 1, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises at least one or two heavy chain variable regions from an antibody described herein, e.g., an antibody chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, or an antibody molecule described in Table 1, or encoded by a nucleotide sequence in Table 1, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.

In some embodiments, the anti-TCRβV antibody molecule comprises a heavy chain variable region (VH) having a consensus sequence of SEQ ID NO: 231 or 3290.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises at least one or two light chain variable regions from an antibody described herein, e.g., an antibody chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, or an antibody described in Table 1, or encoded by a nucleotide sequence in Table 1, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.

In some embodiments, the anti-TCRβV antibody molecule comprises a light chain variable region (VL) having a consensus sequence of SEQ ID NO: 230 or 3289.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises a heavy chain constant region for an IgG4, e.g., a human IgG4. In still another embodiment, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule includes a heavy chain constant region for an IgG1, e.g., a human IgG1. In one embodiment, the heavy chain constant region comprises an amino sequence set forth in Table 3, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) thereto.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, includes a kappa light chain constant region, e.g., a human kappa light chain constant region. In one embodiment, the light chain constant region comprises an amino sequence set forth in Table 3, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) thereto.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, includes at least one, two, or three complementarity determining regions (CDRs) from a heavy chain variable region (VH) of an antibody described herein, e.g., an antibody chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, or an antibody described in Table 1, or encoded by a nucleotide sequence in Table 1, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, includes at least one, two, or three CDRs (or collectively all of the CDRs) from a heavy chain variable region comprising an amino acid sequence shown in Table 1, or encoded by a nucleotide sequence shown in Table 1. In one embodiment, one or more of the CDRs (or collectively all of the CDRs) have one, two, three, four, five, six or more changes, e.g., amino acid substitutions or deletions, relative to the amino acid sequence shown in Table 1, or encoded by a nucleotide sequence shown in Table 1.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, includes at least one, two, or three complementarity determining regions (CDRs) from a light chain variable region of an antibody described herein, e.g., an antibody chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, or an antibody described in Table 1, or encoded by a nucleotide sequence in Table 1, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, includes at least one, two, or three CDRs (or collectively all of the CDRs) from a light chain variable region comprising an amino acid sequence shown in Table 1, or encoded by a nucleotide sequence shown in Table 1. In one embodiment, one or more of the CDRs (or collectively all of the CDRs) have one, two, three, four, five, six or more changes, e.g., amino acid substitutions or deletions, relative to the amino acid sequence shown in Table 1, or encoded by a nucleotide sequence shown in Table 1.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, includes at least one, two, three, four, five or six CDRs (or collectively all of the CDRs) from a heavy and light chain variable region comprising an amino acid sequence shown in Table 1, or encoded by a nucleotide sequence shown in Table 1. In one embodiment, one or more of the CDRs (or collectively all of the CDRs) have one, two, three, four, five, six or more changes, e.g., amino acid substitutions or deletions, relative to the amino acid sequence shown in Table 1, or encoded by a nucleotide sequence shown in Table 1.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, molecule includes all six CDRs from an antibody described herein, e.g., an antibody chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, or an antibody described in Table 1, or encoded by a nucleotide sequence in Table 1, or closely related CDRs, e.g., CDRs which are identical or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions). In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, may include any CDR described herein.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule includes at least one, two, or three CDRs according to Kabat et al. (e.g., at least one, two, or three CDRs according to the Kabat definition as set out in Table 1) from a heavy chain variable region of an antibody described herein, e.g., an antibody chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, or an antibody described in Table 1, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to one, two, or three CDRs according to Kabat et al. shown in Table 1.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule includes at least one, two, or three CDRs according to Kabat et al. (e.g., at least one, two, or three CDRs according to the Kabat definition as set out in Table 1) from a light chain variable region of an antibody described herein, e.g., an antibody chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, or an antibody described in Table 1, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to one, two, or three CDRs according to Kabat et al. shown in Table 1.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, includes at least one, two, three, four, five, or six CDRs according to Kabat et al. (e.g., at least one, two, three, four, five, or six CDRs according to the Kabat definition as set out in Table 1) from the heavy and light chain variable regions of an antibody described herein, e.g., an antibody chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, or an antibody described in Table 1, or encoded by a nucleotide sequence in Table 1; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to one, two, three, four, five, or six CDRs according to Kabat et al. shown in Table 1.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, includes all six CDRs according to Kabat et al. (e.g., all six CDRs according to the Kabat definition as set out in Table 1) from the heavy and light chain variable regions of an antibody described herein, e.g., an antibody chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, or an antibody described in Table 1, or encoded by a nucleotide sequence in Table 1; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to all six CDRs according to Kabat et al. shown in Table 1. In one embodiment, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, may include any CDR described herein.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, includes at least one, two, or three hypervariable loops that have the same canonical structures as the corresponding hypervariable loop of an antibody described herein, e.g., an antibody chosen from chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, e.g., the same canonical structures as at least loop 1 and/or loop 2 of the heavy and/or light chain variable domains of an antibody described herein. See, e.g., Chothia et al., (1992) J. Mol. Biol. 227:799-817; Tomlinson et al., (1992) J. Mol. Biol. 227:776-798 for descriptions of hypervariable loop canonical structures. These structures can be determined by inspection of the tables described in these references.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule includes at least one, two, or three CDRs according to Chothia et al. (e.g., at least one, two, or three CDRs according to the Chothia definition as set out in Table 1) from a heavy chain variable region of an antibody described herein, e.g., an antibody chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, or as described in Table 1, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to one, two, or three CDRs according to Chothia et al. shown in Table 1.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule includes at least one, two, or three CDRs according to Chothia et al. (e.g., at least one, two, or three CDRs according to the Chothia definition as set out in Table 1) from a light chain variable region of an antibody described herein, e.g., an antibody chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, or an antibody described in Table 1, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to one, two, or three CDRs according to Chothia et al. shown in Table 1.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, includes at least one, two, three, four, five, or six CDRs according to Chothia et al. (e.g., at least one, two, three, four, five, or six CDRs according to the Chothia definition as set out in Table 1) from the heavy and light chain variable regions of an antibody described herein, e.g., an antibody chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, or an antibody described in Table 1, or encoded by the nucleotide sequence in Table 1; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to one, two, three, four, five, or six CDRs according to Chothia et al. shown in Table 1.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, includes all six CDRs according to Chothia et al. (e.g., all six CDRs according to the Chothia definition as set out in Table 1) from the heavy and light chain variable regions of an antibody described herein, e.g., an antibody chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, or an antibody described in Table 1, or encoded by a nucleotide sequence in Table 1; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to all six CDRs according to Chothia et al. shown in Table 1. In one embodiment, the anti-TCRβV antibody molecule, e.g., anti-TCRβV6 (e.g., anti-TCRβ V6-5*01) antibody molecule, may include any CDR described herein.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, molecule includes a combination of CDRs or hypervariable loops defined according to Kabat et al., Chothia et al., or as described in Table 1.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, can contain any combination of CDRs or hypervariable loops according to the Kabat and Chothia definitions.

In some embodiments, a combined CDR as set out in Table 1 is a CDR that comprises a Kabat CDR and a Chothia CDR.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, molecule includes a combination of CDRs or hypervariable loops identified as combined CDRs in Table 1. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, can contain any combination of CDRs or hypervariable loops according the “combined” CDRs are described in Table 1.

In an embodiment, e.g., an embodiment comprising a variable region, a CDR (e.g., a combined CDR, Chothia CDR or Kabat CDR), or other sequence referred to herein, e.g., in Table 1, the antibody molecule is a monospecific antibody molecule, a bispecific antibody molecule, a bivalent antibody molecule, a biparatopic antibody molecule, or an antibody molecule that comprises an antigen binding fragment of an antibody, e.g., a half antibody or antigen binding fragment of a half antibody. In certain embodiments the antibody molecule comprises a multispecific molecule, e.g., a bispecific molecule, e.g., as described herein.

In an embodiment, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule includes:

    • (i) one, two or all of a light chain complementarity determining region 1 (LC CDR1), a light chain complementarity determining region 2 (LC CDR2), and a light chain complementarity determining region 3 (LC CDR3) of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11, and/or
    • (ii) one, two or all of a heavy chain complementarity determining region 1 (HC CDR1), heavy chain complementarity determining region 2 (HC CDR2), and a heavy chain complementarity determining region 3 (HC CDR3) of SEQ ID NO: 1 or SEQ ID NO: 9.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule comprises a LC CDR1, LC CDR2, and LC CDR3 of SEQ ID NO: 2, and a HC CDR1, HC CDR2, and HC CDR3 of SEQ ID NO: 1.

In some embodiments the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule comprises a LC CDR1, LC CDR2, and LC CDR3 of SEQ ID NO: 10, and a HC CDR1, HC CDR2, and HC CDR3 of SEQ ID NO: 9.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule comprises a LC CDR1, LC CDR2, and LC CDR3 of SEQ ID NO: 11, and a HC CDR1, HC CDR2, and HC CDR3 of SEQ ID NO: 9.

In an embodiment, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule comprises:

    • (i) a LC CDR1 amino acid sequence of SEQ ID NO: 6, a LC CDR2 amino acid sequence of SEQ ID NO: 7, or a LC CDR3 amino acid sequence of SEQ ID NO: 8; and/or
    • (ii) a HC CDR1 amino acid sequence of SEQ ID NO: 3, a HC CDR2 amino acid sequence of SEQ ID NO: 4, or a HC CDR3 amino acid sequence of SEQ ID NO: 5.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule comprises:

    • (i) a light chain variable region (VL) comprising a LC CDR1 amino acid sequence of SEQ ID NO: 6, a LC CDR2 amino acid sequence of SEQ ID NO: 7, or a LC CDR3 amino acid sequence of SEQ ID NO: 8; and/or
    • (ii) a heavy chain variable region (VH) comprising a HC CDR1 amino acid sequence of SEQ ID NO: 3, a HC CDR2 amino acid sequence of SEQ ID NO: 4, or a HC CDR3 amino acid sequence of SEQ ID NO: 5.

In an embodiment, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule comprises:

    • (i) a LC CDR1 amino acid sequence of SEQ ID NO: 51, a LC CDR2 amino acid sequence of SEQ ID NO: 52, or a LC CDR3 amino acid sequence of SEQ ID NO: 53; and/or
    • (ii) a HC CDR1 amino acid sequence of SEQ ID NO: 45, a HC CDR2 amino acid sequence of SEQ ID NO: 46, or a HC CDR3 amino acid sequence of SEQ ID NO: 47.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule comprises:

    • (i) a light chain variable region (VL) comprising a LC CDR1 amino acid sequence of SEQ ID NO: 51, a LC CDR2 amino acid sequence of SEQ ID NO: 52, or a LC CDR3 amino acid sequence of SEQ ID NO: 53; and/or
    • (ii) a heavy chain variable region (VH) comprising a HC CDR1 amino acid sequence of SEQ ID NO: 45, a HC CDR2 amino acid sequence of SEQ ID NO: 46, or a HC CDR3 amino acid sequence of SEQ ID NO: 47.

In an embodiment, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule comprises:

    • (i) a LC CDR1 amino acid sequence of SEQ ID NO: 54, a LC CDR2 amino acid sequence of SEQ ID NO: 55, or a LC CDR3 amino acid sequence of SEQ ID NO: 56; and/or
    • (ii) a HC CDR1 amino acid sequence of SEQ ID NO: 48, a HC CDR2 amino acid sequence of SEQ ID NO: 49, or a HC CDR3 amino acid sequence of SEQ ID NO: 50.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule comprises:

    • (i) a light chain variable region (VL) comprising a LC CDR1 amino acid sequence of SEQ ID NO: 54, a LC CDR2 amino acid sequence of SEQ ID NO: 55, or a LC CDR3 amino acid sequence of SEQ ID NO: 56; and/or
    • (ii) a heavy chain variable region (VH) comprising a HC CDR1 amino acid sequence of SEQ ID NO: 48, a HC CDR2 amino acid sequence of SEQ ID NO: 49, or a HC CDR3 amino acid sequence of SEQ ID NO: 50.

In one embodiment, the light or the heavy chain variable framework (e.g., the region encompassing at least FR1, FR2, FR3, and optionally FR4) of the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule can be chosen from: (a) a light or heavy chain variable framework including at least 80%, 85%, 87% 90%, 92%, 93%, 95%, 97%, 98%, or 100% of the amino acid residues from a human light or heavy chain variable framework, e.g., a light or heavy chain variable framework residue from a human mature antibody, a human germline sequence, or a human consensus sequence; (b) a light or heavy chain variable framework including from 20% to 80%, 40% to 60%, 60% to 90%, or 70% to 95% of the amino acid residues from a human light or heavy chain variable framework, e.g., a light or heavy chain variable framework residue from a human mature antibody, a human germline sequence, or a human consensus sequence; (c) a non-human framework (e.g., a rodent framework); or (d) a non-human framework that has been modified, e.g., to remove antigenic or cytotoxic determinants, e.g., deimmunized, or partially humanized. In one embodiment, the light or heavy chain variable framework region (particularly FR1, FR2 and/or FR3) includes a light or heavy chain variable framework sequence at least 70, 75, 80, 85, 87, 88, 90, 92, 94, 95, 96, 97, 98, 99% identical or identical to the frameworks of a VL or VH segment of a human germline gene.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises a heavy chain variable domain having at least one, two, three, four, five, six, seven, ten, fifteen, twenty or more changes, e.g., amino acid substitutions or deletions, from an amino acid sequence of any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, e.g., the amino acid sequence of the FR region in the entire variable region, e.g., shown in FIG. 1A, or in SEQ ID NO: 9.

Alternatively, or in combination with the heavy chain substitutions described herein, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises a light chain variable domain having at least one, two, three, four, five, six, seven, ten, fifteen, twenty or more amino acid changes, e.g., amino acid substitutions or deletions, from an amino acid sequence of any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, e.g., the amino acid sequence of the FR region in the entire variable region, e.g., shown in FIG. 1B, or in SEQ ID NO: 10 or SEQ ID NO: 11.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, includes one, two, three, or four heavy chain framework regions shown in FIG. 1A, or a sequence substantially identical thereto.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, includes one, two, three, or four light chain framework regions shown in FIG. 1B, or a sequence substantially identical thereto.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises the light chain framework region 1 of A-H.1 or A-H.2, e.g., as shown in FIG. 1B.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises the light chain framework region 2 of A-H.1 or A-H.2, e.g., as shown in FIG. 1B.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises the light chain framework region 3 of A-H.1 or A-H.2, e.g., as shown in FIG. 1B.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises the light chain framework region 4 of A-H.1 or A-H.2, e.g., as shown in FIG. 1B.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises a light chain variable domain comprising a framework region, e.g., framework region 1 (FR1), comprising a change, e.g., a substitution (e.g., a conservative substitution) at position 10 according to Kabat numbering. In some embodiments, the FR1 comprises a Phenylalanine at position 10, e.g., a Serine to Phenyalanine substitution. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises a light chain variable domain comprising a framework region, e.g., framework region 2 (FR2), comprising a change, e.g., a substitution (e.g., a conservative substitution) at a position disclosed herein according to Kabat numbering. In some embodiments, FR2 comprises a Histidine at position 36, e.g., a substitution at position 36 according to Kabat numbering, e.g., a Tyrosine to Histidine substitution. In some embodiments, FR2 comprises an Alanine at position 46, e.g., a substitution at position 46 according to Kabat numbering, e.g., an Arginine to Alanine substitution. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises a light chain variable domain comprising a framework region, e.g., framework region 3 (FR3), comprising a change, e.g., a substitution (e.g., a conservative substitution) at a position disclosed herein according to Kabat numbering. In some embodiments, FR3 comprises a Phenyalanine at position 87, e.g., a substitution at position 87 according to Kabat numbering, e.g., a Tyrosine to Phenyalanine substitution. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises a light chain variable domain comprising: (a) a framework region 1 (FR1) comprising a Phenylalanine at position 10, e.g., a substitution at position 10 according to Kabat numbering, e.g., a Serine to Phenyalanine substitution; (b) a framework region 2 (FR2) comprising a Histidine at position 36, e.g., a substitution at position 36 according to Kabat numbering, e.g., a Tyrosine to Histidine substitution, and a Alanine at position 46, e.g., a substitution at position 46 according to Kabat numbering, e.g., a Arginine to Alanine substitution; and (c) a framework region 3 (FR3) comprising a Phenylalanine at position 87, e.g., a substitution at position 87 according to Kabat numbering, e.g., a Tyrosine to Phenyalanine substitution, e.g., as shown in the amino acid sequence of SEQ ID NO: 10. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises a light chain variable domain comprising: (a) a framework region 2 (FR2) comprising a Histidine at position 36, e.g., a substitution at position 36 according to Kabat numbering, e.g., a Tyrosine to Histidine substitution, and a Alanine at position 46, e.g., a substitution at position 46 according to Kabat numbering, e.g., a Arginine to Alanine substitution; and (b) a framework region 3 (FR3) comprising a Phenylalanine at position 87, e.g., a substitution at position 87 according to Kabat numbering, e.g., a Tyrosine to Phenyalanine substitution, e.g., as shown in the amino acid sequence of SEQ ID NO: 11. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises a light chain variable domain comprising: (a) a framework region 1 (FR1) comprising a change, e.g., a substitution (e.g., a conservative substitution) at one or more (e.g., all) positions disclosed herein according to Kabat numbering, (b) a framework region 2 (FR2) comprising a change, e.g., a substitution (e.g., a conservative substitution) at one or more (e.g., all) position disclosed herein according to Kabat numbering and (c) a framework region 3 (FR3) comprising a change, e.g., a substitution (e.g., a conservative substitution) at one or more (e.g., all) position disclosed herein according to Kabat numbering. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises the heavy chain framework region 1 of A-H.1 or A-H.2, e.g., as shown in FIG. 1A.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises the heavy chain framework region 2 of A-H.1 or A-H.2, e.g., as shown in FIG. 1A

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises the heavy chain framework region 3 of A-H.1 or A-H.2, e.g., as shown in FIG. 1A.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises the heavy chain framework region 4 of A-H.1 or A-H.2, e.g., as shown in FIG. 1A.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises a heavy chain variable domain comprising a framework region, e.g., framework region 3 (FR3), comprising a change, e.g., a substitution (e.g., a conservative substitution) at a position disclosed herein according to Kabat numbering. In some embodiments, FR3 comprises a Threonine at position 73, e.g., a substitution at position 73 according to Kabat numbering, e.g., a Glutamic Acid to Threonine substitution. In some embodiments, FR3 comprises a Glycine at position 94, e.g., a substitution at position 94 according to Kabat numbering, e.g., an Arginine to Glycine substitution. In some embodiments, the substitution is relative to a human germline heavy chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises a heavy chain variable domain comprising a framework region 3 (FR3) comprising a Threonine at position 73, e.g., a substitution at position 73 according to Kabat numbering, e.g., a Glutamic Acid to Threonine substitution, and a Glycine at position 94, e.g., a substitution at position 94 according to Kabat numbering, e.g., a Arginine to Glycine substitution, e.g., as shown in the amino acid sequence of SEQ ID NO: 10.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises the heavy chain framework regions 1˜4 of A-H.1 or A-H.2, e.g., SEQ ID NO: 9, or as shown in FIGS. 1A and 1B.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises the light chain framework regions 1˜4 of A-H.1, e.g., SEQ ID NO: 10, or as shown in FIGS. 1A and 1B.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises the light chain framework regions 1˜4 of A-H.2, e.g., SEQ ID NO: 11, or as shown in FIGS. 1A and 1B.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises the heavy chain framework regions 1˜4 of A-H.1, e.g., SEQ ID NO: 9; and the light chain framework regions 1˜4 of A-H.1, e.g., SEQ ID NO: 10, or as shown in FIGS. 1A and 1B.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises the heavy chain framework regions 1˜4 of A-H.2, e.g., SEQ ID NO: 9; and the light chain framework regions 1˜4 of A-H.2, e.g., SEQ ID NO: 11, or as shown in FIGS. 1A and 1B.

In some embodiments, the heavy or light chain variable domain, or both, of the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, includes an amino acid sequence, which is substantially identical to an amino acid disclosed herein, e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical to a variable region of an antibody described herein, e.g., an antibody chosen from any one of A-H.1 to A-H.85, e.g., A-H.1, A-H.2 or A-H.68, or as described in Table 1, or encoded by the nucleotide sequence in Table 1; or which differs at least 1 or 5 residues, but less than 40, 30, 20, or 10 residues, from a variable region of an antibody described herein.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises at least one, two, three, or four antigen-binding regions, e.g., variable regions, having an amino acid sequence as set forth in Table 1, or a sequence substantially identical thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, or which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the sequences shown in Table 1. In another embodiment, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule includes a VH and/or VL domain encoded by a nucleic acid having a nucleotide sequence as set forth in Table 1, or a sequence substantially identical thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, or which differs by no more than 3, 6, 15, 30, or 45 nucleotides from the sequences shown in Table 1.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 9, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence of SEQ ID NO: 9, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 9; and/or
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 10, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence of SEQ ID NO: 10, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 10.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 9, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence of SEQ ID NO: 9, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 9; and/or
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 11, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence of SEQ ID NO: 11, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 11.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule is a full antibody or fragment thereof (e.g., a Fab, F(ab′)2, Fv, or a single chain Fv fragment (scFv)). In embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule is a monoclonal antibody or an antibody with single specificity. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, can also be a humanized, chimeric, camelid, shark, or an in vitro-generated antibody molecule. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, is a humanized antibody molecule. The heavy and light chains of the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, can be full-length (e.g., an antibody can include at least one, and preferably two, complete heavy chains, and at least one, and preferably two, complete light chains) or can include an antigen-binding fragment (e.g., a Fab, F(ab′)2, Fv, a single chain Fv fragment, a single domain antibody, a diabody (dAb), a bivalent antibody, or bispecific antibody or fragment thereof, a single domain variant thereof, or a camelid antibody).

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, is in the form of a multispecific molecule, e.g., a bispecific molecule, e.g., as described herein.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, has a heavy chain constant region (Fc) chosen from, e.g., the heavy chain constant regions of IgG1, IgG2, IgG3, IgG4, IgM, IgA1, IgA2, IgD, and IgE. In some embodiments, the Fc region is chosen from the heavy chain constant regions of IgG1, IgG2, IgG3, and IgG4. In some embodiments, the Fc region is chosen from the heavy chain constant region of IgG1 or IgG2 (e.g., human IgG1, or IgG2). In some embodiments, the heavy chain constant region is human IgG1. In some embodiments, the Fc region comprises a Fc region variant, e.g., as described herein.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule, has a light chain constant region chosen from, e.g., the light chain constant regions of kappa or lambda, preferably kappa (e.g., human kappa). In one embodiment, the constant region is altered, e.g., mutated, to modify the properties of the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule (e.g., to increase or decrease one or more of: Fc receptor binding, antibody glycosylation, the number of cysteine residues, effector cell function, or complement function). For example, the constant region is mutated at positions 296 (M to Y), 298 (S to T), 300 (T to E), 477 (H to K) and 478 (N to F) to alter Fc receptor binding (e.g., the mutated positions correspond to positions 132 (M to Y), 134 (S to T), 136 (T to E), 313 (H to K) and 314 (N to F) of SEQ ID NOs: 212 or 214; or positions 135 (M to Y), 137 (S to T), 139 (T to E), 316 (H to K) and 317 (N to F) of SEQ ID NOs: 215, 216, 217 or 218), e.g., relative to human IgG1.

Antibody A-H.1 comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 3278 and a light chain comprising the amino acid sequence of SEQ ID NO: 72. Antibody A-H.2 comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 3278 and a light chain comprising the amino acid sequence of SEQ ID NO: 3279. Antibody A-H.68 comprises the amino acid sequence of SEQ ID NO: 1337, or a sequence having at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto. Antibody A-H.69 comprises the amino acid sequence of SEQ ID NO: 1500, or a sequence having at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto.

Additional exemplary humanized anti-TCRB V6 antibodies are provided in Table 1. In some embodiments, the anti-TCRβ V6 is antibody A, e.g., humanized antibody A (antibody A-H), as provided in Table 1. In some embodiments, the anti-TCRβV antibody comprises one or more (e.g., all three) of a LC CDR1, LC CDR2, and LC CDR3 provided in Table 1; and/or one or more (e.g., all three) of a HC CDR1, HC CDR2, and HC CDR3 provided in Table 1, or a sequence with at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto. In some embodiments, antibody A comprises a variable heavy chain (VH) and/or a variable light chain (VL) provided in Table 1, or a sequence with at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule comprises a VH of A-H.1, A-H.2, A-H.3, A-H.4, A-H.5, A-H.6, A-H.7, A-H.8, A-H.9, A-H.10, A-H.11, A-H.12, A-H.13, A-H.14, A-H.15, A-H.16, A-H.17, A-H.18, A-H.19, A-H.20, A-H.21, A-H.22, A-H.23, A-H.24, A-H.25, A-H.26, A-H.27, A-H.28, A-H.29, A-H.30, A-H.31, A-H.32, A-H.33, A-H.34, A-H.35, A-H.36, A-H.37, A-H.38, A-H.39, A-H.40, A-H.1, A-H.42, A-H.43, A-H.44, A-H.45, A-H.46, A-H.47, A-H.48, A-H.49, A-H.50, A-H.51, A-H.52, A-H.53, A-H.54, A-H.55, A-H.56, A-H.57, A-H.58, A-H.59, A-H.60, A-H.61, A-H.62, A-H.63, A-H.64, A-H.65, A-H.66, A-H.67, A-H.68, A-H.69, A-H.70, A-H.71, A-H.72, A-H.73, A-H.74, A-H.75, A-H.76, A-H.77, A-H.78, A-H.79, A-H.80, A-H.81, A-H.82, A-H.83, A-H.84, or A-H.85, or a sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule comprises a VL of A-H.1, A-H.2, A-H.3, A-H.4, A-H.5, A-H.6, A-H.7, A-H.8, A-H.9, A-H.10, A-H.11, A-H.12, A-H.13, A-H.14, A-H.15, A-H.16, A-H.17, A-H.18, A-H.19, A-H.20, A-H.21, A-H.22, A-H.23, A-H.24, A-H.25, A-H.26, A-H.27, A-H.28, A-H.29, A-H.30, A-H.31, A-H.32, A-H.33, A-H.34, A-H.35, A-H.36, A-H.37, A-H.38, A-H.39, A-H.40, A-H.1, A-H.42, A-H.43, A-H.44, A-H.45, A-H.46, A-H.47, A-H.48, A-H.49, A-H.50, A-H.51, A-H.52, A-H.53, A-H.54, A-H.55, A-H.56, A-H.57, A-H.58, A-H.59, A-H.60, A-H.61, A-H.62, A-H.63, A-H.64, A-H.65, A-H.66, A-H.67, A-H.68, A-H.69, A-H.70, A-H.71, A-H.72, A-H.73, A-H.74, A-H.75, A-H.76, A-H.77, A-H.78, A-H.79, A-H.80, A-H.81, A-H.82, A-H.83, A-H.84, or A-H.85, or a sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule comprises a VH of A-H.1, A-H.2, A-H.3, A-H.4, A-H.5, A-H.6, A-H.7, A-H.8, A-H.9, A-H.10, A-H.11, A-H.12, A-H.13, A-H.14, A-H.15, A-H.16, A-H.17, A-H.18, A-H.19, A-H.20, A-H.21, A-H.22, A-H.23, A-H.24, A-H.25, A-H.26, A-H.27, A-H.28, A-H.29, A-H.30, A-H.31, A-H.32, A-H.33, A-H.34, A-H.35, A-H.36, A-H.37, A-H.38, A-H.39, A-H.40, A-H.1, A-H.42, A-H.43, A-H.44, A-H.45, A-H.46, A-H.47, A-H.48, A-H.49, A-H.50, A-H.51, A-H.52, A-H.53, A-H.54, A-H.55, A-H.56, A-H.57, A-H.58, A-H.59, A-H.60, A-H.61, A-H.62, A-H.63, A-H.64, A-H.65, A-H.66, A-H.67, A-H.68, A-H.69, A-H.70, A-H.71, A-H.72, A-H.73, A-H.74, A-H.75, A-H.76, A-H.77, A-H.78, A-H.79, A-H.80, A-H.81, A-H.82, A-H.83, A-H.84, or A-H.85, or a sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto; and a VL of A-H.1, A-H.2, A-H.3, A-H.4, A-H.5, A-H.6, A-H.7, A-H.8, A-H.9, A-H.10, A-H.11, A-H.12, A-H.13, A-H.14, A-H.15, A-H.16, A-H.17, A-H.18, A-H.19, A-H.20, A-H.21, A-H.22, A-H.23, A-H.24, A-H.25, A-H.26, A-H.27, A-H.28, A-H.29, A-H.30, A-H.31, A-H.32, A-H.33, A-H.34, A-H.35, A-H.36, A-H.37, A-H.38, A-H.39, A-H.40, A-H.1, A-H.42, A-H.43, A-H.44, A-H.45, A-H.46, A-H.47, A-H.48, A-H.49, A-H.50, A-H.51, A-H.52, A-H.53, A-H.54, A-H.55, A-H.56, A-H.57, A-H.58, A-H.59, A-H.60, A-H.61, A-H.62, A-H.63, A-H.64, A-H.65, A-H.66, A-H.67, A-H.68, A-H.69, A-H.70, A-H.71, A-H.72, A-H.73, A-H.74, A-H.75, A-H.76, A-H.77, A-H.78, A-H.79, A-H.80, A-H.81, A-H.82, A-H.83, A-H.84, or A-H.85, or a sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.

TABLE 1 Amino acid and nucleotide sequences for murine, chimeric and humanized antibody molecules which bind to TCRVB 6, e.g.,   TCRVB 6-5. The antibody molecules include murine mAb Antibody A,  and humanized mAb Antibody A-H Clones A-H.1 to A-H.85. The amino acid the heavy and light chain CDRs, and the amino acid  and nucleotide sequences of the heavy and light chain variable  regions, and the heavy and light chains are shown. Antibody A (murine), also referred to as H131, TCRVB 6-5 binder SEQ ID NO: 3 HC CDR1 (Combined) GYSFTTYYIH SEQ ID NO: 4 HC CDR2 (Combined) WFFPGSGNIKYNEKFKG SEQ ID NO: 5 HC CDR3 (Combined) SYYSYDVLDY SEQ ID NO: 45 HC CDR1 (Kabat) TYYIH SEQ ID NO: 46 HC CDR2 (Kabat) WFFPGSGNIKYNEKFKG SEQ ID NO: 47 HC CDR3 (Kabat) SYYSYDVLDY SEQ ID NO: 48 HC CDR1 (Chothia) GYSFTTY SEQ ID NO: 49 HC CDR2 (Chothia) FPGSGN SEQ ID NO: 50 HC CDR3 (Chothia) SYYSYDVLDY SEQ ID NO: 1 VH QVQLQQSGPELVKPGTSVKISCKASGYSFTTYYIHW VKQRPGQGLEWIGWFFPGSGNIKYNEKFKGKATLT ADTSSSTAYMQLSSLTSEESAVYFCAGSYYSYDVLD YWGHGTTLTVSS SEQ ID NO: 6 LC CDR1 (Combined) KASQNVGINVV SEQ ID NO: 7 LC CDR2 (Combined) SSSHRYS SEQ ID NO: 8 LC CDR3 (Combined) QQFKSYPLT SEQ ID NO: 51 LC CDR1 (Kabat) KASQNVGINVV SEQ ID NO: 52 LC CDR2 (Kabat) SSSHRYS SEQ ID NO: 53 LC CDR3 (Kabat) QQFKSYPLT SEQ ID NO: 54 LC CDR1 (Chothia) KASQNVGINVV SEQ ID NO: 55 LC CDR2 (Chothia) SSSHRYS SEQ ID NO: 56 LC CDR3 (Chothia) QQFKSYPLT SEQ ID NO: 2 VL DILMTQSQKFMSTSLGDRVSVSCKASQNVGINVVW HQQKPGQSPKALIYSSSHRYSGVPDRFTGSGSGTDFT LTINNVQSEDLAEYFCQQFKSYPLTFGAGTKLELK Antibody A humanized (A-H antibody), TCRVB 6-5 binder A-H.1 antibody (also referred to as BHM1709) SEQ ID NO: 3 HC CDR1 (Combined) GYSFTTYYIH SEQ ID NO: 4 HC CDR2 (Combined) WFFPGSGNIKYNEKFKG SEQ ID NO: 5 HC CDR3 (Combined) SYYSYDVLDY SEQ ID NO: 9 VH QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFFPGSGNIKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS SEQ ID NO: 12 DNA VH CAGGTGCAGCTGGTTCAGTCTGGCGCCGAAGTGA AGAAACCTGGCTCCTCCGTGAAGGTGTCCTGCAA GGCTTCCGGCTACTCCTTCACCACCTACTACATCC ACTGGGTCCGACAGGCCCCTGGACAAGGATTGGA ATGGATGGGCTGGTTCTTCCCCGGCTCCGGCAACA TCAAGTACAACGAGAAGTTCAAGGGCCGCGTGAC CATCACCGCCGACACCTCTACCTCTACCGCCTACA TGGAACTGTCCAGCCTGAGATCTGAGGACACCGC CGTGTACTACTGCGCCGGCTCCTACTACTCTTACG ACGTGCTGGATTACTGGGGCCAGGGCACCACAGT GACAGTGTCCTCT SEQ ID NO: 69 VH-IgM constant delta METDTLLLWVLLLWVPGSTGQVQLVQSGAEVKKP CDC GSSVKVSCKASGYSFTTYYIHWVRQAPGQGLEWMG WFFPGSGNIKYNEKFKGRVTITADTSTSTAYMELSSL RSEDTAVYYCAGSYYSYDVLDYWGQGTTVTVSSGS ASAPTLFPLVSCENSPSDTSSVAVGCLAQDFLPDSITF SWKYKNNSDISSTRGFPSVLRGGKYAATSQVLLPSK DVMQGTDEHVVCKVQHPNGNKEKNVPLPVIAELPP KVSVFVPPRDGFFGNPRKSKLICQATGFSPRQIQVSW LREGKQVGSGVTTDQVQAEAKESGPTTYKVTSTLTI KESDWLGQSMFTCRVDHRGLTFQQNASSMCVPDQ DTAIRVFAIPPSFASIFLTKSTKLTCLVTDLTTYDSVTI SWTRQNGEAVKTHTNISESHPNATFSAVGEASICED DWNSGERFTCTVTHTDLASSLKQTISRPKGVALHRP DVYLLPPAREQLNLRESATITCLVTGFSPADVFVQW MQRGQPLSPEKYVTSAPMPEPQAPGRYFAHSILTVS EEEWNTGETYTCVVAHEALPNRVTERTVDKSTGKP TLYNVSLVMSDTAGTCY SEQ ID NO: 70 VH-IgGA1 METDTLLLWVLLLWVPGSTGQVQLVQSGAEVKKP GSSVKVSCKASGYSFTTYYIHWVRQAPGQGLEWMG WFFPGSGNIKYNEKFKGRVTITADTSTSTAYMELSSL RSEDTAVYYCAGSYYSYDVLDYWGQGTTVTVSSAS PTSPKVFPLSLCSTQPDGNVVIACLVQGFFPQEPLSV TWSESGQGVTARNFPPSQDASGDLYTTSSQLTLPAT QCLAGKSVTCHVKHYTNPSQDVTVPCPVPSTPPTPS PSTPPTPSPSCCHPRLSLHRPALEDLLLGSEANLTCTL TGLRDASGVTFTWTPSSGKSAVQGPPERDLCGCYSV SSVLPGCAEPWNHGKTFTCTAAYPESKTPLTATLSK SGNTFRPEVHLLPPPSEELALNELVTLTCLARGFSPK DVLVRWLQGSQELPREKYLTWASRQEPSQGTTTFA VTSILRVAAEDWKKGDTFSCMVGHEALPLAFTQKTI DRLAGKPTHVNVSVVMAEVDGTCY SEQ ID NO: 71 VH-IgGA2 METDTLLLWVLLLWVPGSTGQVQLVQSGAEVKKP GSSVKVSCKASGYSFTTYYIHWVRQAPGQGLEWMG WFFPGSGNIKYNEKFKGRVTITADTSTSTAYMELSSL RSEDTAVYYCAGSYYSYDVLDYWGQGTTVTVSSAS PTSPKVFPLSLDSTPQDGNVVVACLVQGFFPQEPLSV TWSESGQNVTARNFPPSQDASGDLYTTSSQLTLPAT QCPDGKSVTCHVKHYTNSSQDVTVPCRVPPPPPCCH PRLSLHRPALEDLLLGSEANLTCTLTGLRDASGATFT WTPSSGKSAVQGPPERDLCGCYSVSSVLPGCAQPW NHGETFTCTAAHPELKTPLTANITKSGNTFRPEVHLL PPPSEELALNELVTLTCLARGFSPKDVLVRWLQGSQ ELPREKYLTWASRQEPSQGTTTYAVTSILRVAAEDW KKGETFSCMVGHEALPLAFTQKTIDRMAGKPTHINV SVVMAEADGTCY SEQ ID NO: Heavy chain METDTLLLWVLLLWVPGSTGQVQLVQSGAEVKKP 3278 GSSVKVSCKASGYSFTTYYIHWVRQAPGQGLEWMG WFFPGSGNIKYNEKFKGRVTITADTSTSTAYMELSSL RSEDTAVYYCAGSYYSYDVLDYWGQGTTVTVSSAS TKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV SWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSL GTQTYICNVNHKPSNTKVDKRVEPKSCDKTHTCPPC PAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDV SHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYR VVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS KAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGF YPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYS KLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSL SPGK SEQ ID NO: 6 LC CDR1 (Combined) KASQNVGINVV SEQ ID NO: 7 LC CDR2 (Combined) SSSHRYS SEQ ID NO: 8 LC CDR3 (Combined) QQFKSYPLT SEQ ID NO: 10 VL DIQMTQSPSFLSASVGDRVTITCKASQNVGINVVWH QQKPGKAPKALIYSSSHRYSGVPSRFSGSGSGTEFTL TISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 13 DNA VL GACATCCAGATGACCCAGTCTCCATCCTTCCTGTC CGCCTCTGTGGGCGACAGAGTGACCATCACATGC AAGGCCTCTCAGAACGTGGGCATCAACGTCGTGT GGCACCAGCAGAAGCCTGGCAAGGCTCCTAAGGC TCTGATCTACTCCTCCAGCCACCGGTACTCTGGCG TGCCCTCTAGATTTTCCGGCTCTGGCTCTGGCACC GAGTTTACCCTGACAATCTCCAGCCTGCAGCCTGA GGACTTCGCCACCTACTTTTGCCAGCAGTTCAAGA GCTACCCTCTGACCTTTGGCCAGGGCACCAAGCTG GAAATCAAG SEQ ID NO: 72 VL and kappa constant METDTLLLWVLLLWVPGSTGDIQMTQSPSFLSASVG region/light chain DRVTITCKASQNVGINVVWHQQKPGKAPKALIYSSS HRYSGVPSRFSGSGSGTEFTLTISSLQPEDFATYFCQ QFKSYPLTFGQGTKLEIKRTVAAPSVFIFPPSDEQLKS GTASVVCLLNNFYPREAKVQWKVDNALQSGNSQES VTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTH QGLSSPVTKSFNRGEC A-H.2 antibody (also referred to as BHM1710) SEQ ID NO: 3 HC CDR1 (Combined) GYSFTTYYIH SEQ ID NO: 4 HC CDR2 (Combined) WFFPGSGNIKYNEKFKG SEQ ID NO: 5 HC CDR3 (Combined) SYYSYDVLDY SEQ ID NO: 9 VH QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFFPGSGNIKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS SEQ ID NO: 12 DNA VH CAGGTGCAGCTGGTTCAGTCTGGCGCCGAAGTGA AGAAACCTGGCTCCTCCGTGAAGGTGTCCTGCAA GGCTTCCGGCTACTCCTTCACCACCTACTACATCC ACTGGGTCCGACAGGCCCCTGGACAAGGATTGGA ATGGATGGGCTGGTTCTTCCCCGGCTCCGGCAACA TCAAGTACAACGAGAAGTTCAAGGGCCGCGTGAC CATCACCGCCGACACCTCTACCTCTACCGCCTACA TGGAACTGTCCAGCCTGAGATCTGAGGACACCGC CGTGTACTACTGCGCCGGCTCCTACTACTCTTACG ACGTGCTGGATTACTGGGGCCAGGGCACCACAGT GACAGTGTCCTCT SEQ ID NO: Heavy chain METDTLLLWVLLLWVPGSTGQVQLVQSGAEVKKP 3278 GSSVKVSCKASGYSFTTYYIHWVRQAPGQGLEWMG WFFPGSGNIKYNEKFKGRVTITADTSTSTAYMELSSL RSEDTAVYYCAGSYYSYDVLDYWGQGTTVTVSSAS TKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTV SWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSL GTQTYICNVNHKPSNTKVDKRVEPKSCDKTHTCPPC PAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDV SHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYR VVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS KAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGF YPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYS KLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSL SPGK SEQ ID NO: 6 LC CDR1 (Combined) KASQNVGINVV SEQ ID NO: 7 LC CDR2 (Combined) SSSHRYS SEQ ID NO: 8 LC CDR3 (Combined) QQFKSYPLT SEQ ID NO: 11 VL DIQMTQSPSSLSASVGDRVTITCKASQNVGINVVWH QQKPGKVPKALIYSSSHRYSGVPSRFSGSGSGTDFTL TISSLQPEDVATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 14 DNA VL GACATCCAGATGACCCAGTCTCCATCCTCTCTGTC CGCCTCTGTGGGCGACAGAGTGACCATCACATGC AAGGCCTCTCAGAACGTGGGCATCAACGTCGTGT GGCACCAGCAGAAACCTGGCAAGGTGCCCAAGGC TCTGATCTACTCCTCCAGCCACAGATACTCCGGCG TGCCCTCTAGATTCTCCGGCTCTGGCTCTGGCACC GACTTTACCCTGACAATCTCCAGCCTGCAGCCTGA GGACGTGGCCACCTACTTTTGCCAGCAGTTCAAG AGCTACCCTCTGACCTTTGGCCAGGGCACCAAGCT GGAAATCAAG SEQ ID NO: Light chain METDTLLLWVLLLWVPGSTGDIQMTQSPSSLSASVG 3279 DRVTITCKASQNVGINVVWHQQKPGKVPKALIYSSS HRYSGVPSRFSGSGSGTDFTLTISSLQPEDVATYFCQ QFKSYPLTFGQGTKLEIKRTVAAPSVFIFPPSDEQLKS GTASVVCLLNNFYPREAKVQWKVDNALQSGNSQES VTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTH QGLSSPVTKSFNRGEC A-H.3 antibody SEQ ID NO: 80 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRVSPGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVEDRVAWY QQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTL TISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 81 VL DIQMTQSPSFLSASVGDRVTITCKASQNVEDRVAWY QQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTL TISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 82 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRVSPGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.4 SEQ ID NO: 83 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKIYIH WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVEDRVAWY QQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTL TISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 84 VL DIQMTQSPSFLSASVGDRVTITCKASQNVEDRVAWY QQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTL TISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 85 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKIYIH WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.5 SEQ ID NO: 86 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGHDFRDFYIH WVRQAPGQGLEWMGRVYPGSGSYRYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 87 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 88 VH QVQLVQSGAEVKKPGSSVKVSCKASGHDFRDFYIH WVRQAPGQGLEWMGRVYPGSGSYRYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.6 SEQ ID NO: 89 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGHDFKLTYIH WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVDNRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 90 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDNRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 91 VH QVQLVQSGAEVKKPGSSVKVSCKASGHDFKLTYIH WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.7 SEQ ID NO: 92 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRIFPGSGNVKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVENKVAWHQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 93 VL DIQMTQSPSFLSASVGDRVTITCKASQNVENKVAWH QQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTL TISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 94 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRIFPGSGNVKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.8 SEQ ID NO: 95 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKIYIH WVRQAPGQGLEWMGRIFAGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVDDRVAWYQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 96 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 97 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKIYIH WVRQAPGQGLEWMGRIFAGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.9 SEQ ID NO: 98 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGHDFDKFYIH WVRQAPGQGLEWMGRVSAGSGNVKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYD VLDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGG SDIQMTQSPSFLSASVGDRVTITCKASQNVGNRVAW YQQKPGKAPKALIYSSSHRYSGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 99 VL DIQMTQSPSFLSASVGDRVTITCKASQNVGNRVAW YQQKPGKAPKALIYSSSHRYSGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 100 VH QVQLVQSGAEVKKPGSSVKVSCKASGHDFDKFYIH WVRQAPGQGLEWMGRVSAGSGNVKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYD VLDYWGQGTTVTVSSS A-H.10 SEQ ID NO: 101 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGHDFDKFYIH WVRQAPGQGLEWMGRIFAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVGDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIKs SEQ ID NO: 102 VL DIQMTQSPSFLSASVGDRVTITCKASQNVGDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 103 VH QVQLVQSGAEVKKPGSSVKVSCKASGHDFDKFYIH WVRQAPGQGLEWMGRIFAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.11 SEQ ID NO: 104 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRVSPGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVGDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 105 VL DIQMTQSPSFLSASVGDRVTITCKASQNVGDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 106 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRVSPGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.12 SEQ ID NO: 107 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKIYIH WVRQAPGQGLEWMGRVSAGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVGNRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 108 VL DIQMTQSPSFLSASVGDRVTITCKASQNVGNRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 109 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKIYIH WVRQAPGQGLEWMGRVSAGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.13, also referred to as A-H.69 SEQ ID NO: 110 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRIFPGSGNVKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVDNRVAWYQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 111 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDNRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 112 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRIFPGSGNVKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.14 SEQ ID NO: 113 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKIYIH WVRQAPGQGLEWMGRISAGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVDDRVAWYQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 114 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 115 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKIYIH WVRQAPGQGLEWMGRISAGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.15 SEQ ID NO: 116 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFRLTYIH WVRQAPGQGLEWMGRVSPGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVDNKVAW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 117 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDNKVAW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 118 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFRLTYIH WVRQAPGQGLEWMGRVSPGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.16 SEQ ID NO: 119 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGGTFRLTYIH WVRQAPGQGLEWMGRVYPGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 120 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 121 VH QVQLVQSGAEVKKPGSSVKVSCKASGGTFRLTYIH WVRQAPGQGLEWMGRVYPGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.17 SEQ ID NO: 122 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFRLTYIH WVRQAPGQGLEWMGRIFPGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVDDRVAWYQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 123 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 124 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFRLTYIH WVRQAPGQGLEWMGRIFPGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.18 SEQ ID NO: 125 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRIFPGSGNVKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVEDRVAWYQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 126 VL DIQMTQSPSFLSASVGDRVTITCKASQNVEDRVAWY QQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTL TISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 127 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRIFPGSGNVKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.19 SEQ ID NO: 128 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGGTFRLTYIH WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVGDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 129 VL DIQMTQSPSFLSASVGDRVTITCKASQNVGDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 130 VH QVQLVQSGAEVKKPGSSVKVSCKASGGTFRLTYIH WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.20 SEQ ID NO: 131 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGGTFDKTYIH WVRQAPGQGLEWMGRISAGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVDDRVAWYQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 132 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 133 VH QVQLVQSGAEVKKPGSSVKVSCKASGGTFDKTYIH WVRQAPGQGLEWMGRISAGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.21 SEQ ID NO: 134 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGHDFDKFYIH WVRQAPGQGLEWMGRISAGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVDDRVAWYQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 135 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 136 VH QVQLVQSGAEVKKPGSSVKVSCKASGHDFDKFYIH WVRQAPGQGLEWMGRISAGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.22 SEQ ID NO: 137 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVDNKVAW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 138 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDNKVAW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 139 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.23 SEQ ID NO: 140 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGHDFRLTYIH WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVADRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 141 VL DIQMTQSPSFLSASVGDRVTITCKASQNVADRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 142 VH QVQLVQSGAEVKKPGSSVKVSCKASGHDFRLTYIH WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.24 SEQ ID NO: 143 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGHDFHLWYIH WVRQAPGQGLEWMGRVSAGSGNVKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYD VLDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGG SDIQMTQSPSFLSASVGDRVTITCKASQNVDNKVAW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 144 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDNKVAW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 145 VH QVQLVQSGAEVKKPGSSVKVSCKASGHDFHLWYIH WVRQAPGQGLEWMGRVSAGSGNVKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYD VLDYWGQGTTVTVSS A-H.25 SEQ ID NO: 146 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGHDFHLWYIH WVRQAPGQGLEWMGRVFAGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVEDKVAWY QQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTL TISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 147 VL DIQMTQSPSFLSASVGDRVTITCKASQNVEDKVAWY QQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTL TISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 148 VH QVQLVQSGAEVKKPGSSVKVSCKASGHDFHLWYIH WVRQAPGQGLEWMGRVFAGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.26 SEQ ID NO: 149 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRIFPGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVDDRVAWYQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 150 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 151 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRIFPGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.27 SEQ ID NO: 153 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRVSAGSGNVKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYD VLDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGG SDIQMTQSPSFLSASVGDRVTITCKASQNVGNRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 154 VL DIQMTQSPSFLSASVGDRVTITCKASQNVGNRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 155 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRVSAGSGNVKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYD VLDYWGQGTTVTVSS A-H.28 SEQ ID NO: 156 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRISPGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVGDRVAWYQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 157 VL DIQMTQSPSFLSASVGDRVTITCKASQNVGDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 158 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRISPGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.29 SEQ ID NO: 159 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGHDFHLWYIH WVRQAPGQGLEWMGRISPGSGNVKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVGDRVAWHQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 160 VL DIQMTQSPSFLSASVGDRVTITCKASQNVGDRVAW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 161 VH QVQLVQSGAEVKKPGSSVKVSCKASGHDFHLWYIH WVRQAPGQGLEWMGRISPGSGNVKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.31 SEQ ID NO: 162 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGHDFKLTYIH WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 163 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 164 VH QVQLVQSGAEVKKPGSSVKVSCKASGHDFKLTYIH WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.31 SEQ ID NO: 165 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFHLWYIH WVRQAPGQGLEWMGRVFAGSGSYRYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 166 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 167 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFHLWYIH WVRQAPGQGLEWMGRVFAGSGSYRYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.32 SEQ ID NO: 168 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKIYIH WVRQAPGQGLEWMGRISAGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVADRVAWYQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 169 VL DIQMTQSPSFLSASVGDRVTITCKASQNVADRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 170 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKIYIH WVRQAPGQGLEWMGRISAGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.33 SEQ ID NO: 171 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRISAGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVEDRVAWYQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 172 VL DIQMTQSPSFLSASVGDRVTITCKASQNVEDRVAWY QQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTL TISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 173 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRISAGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.34 SEQ ID NO: 174 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFRLTYIH WVRQAPGQGLEWMGRISPGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVGNRVAWYQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 175 VL DIQMTQSPSFLSASVGDRVTITCKASQNVGNRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 176 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFRLTYIH WVRQAPGQGLEWMGRISPGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.35 SEQ ID NO: 177 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGHDFDKTYIH WVRQAPGQGLEWMGRVSAGSGNVKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYD VLDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGG SDIQMTQSPSFLSASVGDRVTITCKASQNVEDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 178 VL DIQMTQSPSFLSASVGDRVTITCKASQNVEDRVAWY QQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTL TISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 179 VH QVQLVQSGAEVKKPGSSVKVSCKASGHDFDKTYIH WVRQAPGQGLEWMGRVSAGSGNVKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYD VLDYWGQGTTVTVSS A-H.36 SEQ ID NO: 180 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGHDFKLTYIH WVRQAPGQGLEWMGRVSPGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVEDRVAWH QQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTL TISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 181 VL DIQMTQSPSFLSASVGDRVTITCKASQNVEDRVAWH QQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTL TISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 182 VH QVQLVQSGAEVKKPGSSVKVSCKASGHDFKLTYIH WVRQAPGQGLEWMGRVSPGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.37 SEQ ID NO: 183 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGHDFDKTYIH WVRQAPGQGLEWMGRIYPGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVADRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 184 VL DIQMTQSPSFLSASVGDRVTITCKASQNVADRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 185 VH QVQLVQSGAEVKKPGSSVKVSCKASGHDFDKTYIH WVRQAPGQGLEWMGRIYPGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.38 SEQ ID NO: 186 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKTYIH WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 187 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 188 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKTYIH WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.39 SEQ ID NO: 189 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKIYIH WVRQAPGQGLEWMGRISAGSGNIKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVDDRVAWYQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 190 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 191 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKIYIH WVRQAPGQGLEWMGRISAGSGNIKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.40 SEQ ID NO: 192 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKIYIH WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVGDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 193 VL DIQMTQSPSFLSASVGDRVTITCKASQNVGDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 194 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKIYIH WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.41 SEQ ID NO: 195 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGGTFKLTYIH WVRQAPGQGLEWMGRVSAGSGNVKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYD VLDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGG SDIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 196 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDDRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 197 VH QVQLVQSGAEVKKPGSSVKVSCKASGGTFKLTYIH WVRQAPGQGLEWMGRVSAGSGNVKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYD VLDYWGQGTTVTVSS A-H.42 SEQ ID NO: 198 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRISPGSGNVKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVDNRVAWHQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 199 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDNRVAW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 200 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRISPGSGNVKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.43 SEQ ID NO: 201 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGHDFDKFYIH WVRQAPGQGLEWMGRVSAGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVDNRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 202 VL DIQMTQSPSFLSASVGDRVTITCKASQNVDNRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 203 VH QVQLVQSGAEVKKPGSSVKVSCKASGHDFDKFYIH WVRQAPGQGLEWMGRVSAGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.44 SEQ ID NO: 204 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKFYIH WVRQAPGQGLEWMGRVSAGSGNVKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYD VLDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGG SDIQMTQSPSFLSASVGDRVTITCKASQNVGDRVVW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 205 VH QVQLVQSGAEVKKPGSSVKVSCKASGTDFDKFYIH WVRQAPGQGLEWMGRVSAGSGNVKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYD VLDYWGQGTTVTVSS A-H.45 SEQ ID NO: 206 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFSAGSGNTKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYD VLDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGG SDIQMTQSPSFLSASVGDRVTITCKASQNVGINVVW HQQKPGKAPKALIYSSSHRYSGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 207 VH QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFSAGSGNTKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYD VLDYWGQGTTVTVSS A-H.46 SEQ ID NO: 208 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFSAGSGNTKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYD VLDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGG SDIQMTQSPSFLSASVGDRVTITCKASQNVGINVVW HQQKPGKAPKALIYSSSHRYSGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 209 VH QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYH WVRQAPGQGLEWMGWFSAGSGNTKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYD VLDYWGQGTTVTVSS A-H.47 SEQ ID NO: 210 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFFPGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVGINVVWH QQKPGKAPKALIYSSSHRYSGVPSRFSGSGSGTEFTL TISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 211 VH QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFFPGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.48 SEQ ID NO: 212 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFSPGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVGINVVWH QQKPGKAPKALIYSSSHRYSGVPSRFSGSGSGTEFTL TISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 213 VH QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFSPGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYDV LDYWGQGTTVTVSS A-H.49 SEQ ID NO: 214 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFSPGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVGINVVWH QQKPGKAPKALIYSSSHRYSGVPSRFSGSGSGTEFTL TISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 215 VH QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFSPGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDV LDYWGQGTTVTVSS A-H.50 SEQ ID NO: 216 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGRIFPGSGNIKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVGINVVWHQ QKPGKAPKALIYSSSHRYSGVPSRFSGSGSGTEFTLTI SSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 217 VH QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGRIFPGSGNIKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.51 SEQ ID NO: 218 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFFPGSGNIKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSIYSAGVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVGINVVWHQ QKPGKAPKALIYSSSHRYSGVPSRFSGSGSGTEFTLTI SSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 219 VH QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFFPGSGNIKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSIYSAGVL DYWGQGTTVTVSS A-H.52 SEQ ID NO: 220 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFFPGSGNIKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVGINVVWHQ QKPGKAPKALIYSSSHRYSGVPSRFSGSGSGTEFTLTI SSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 221 VH QVQLVQSGAEVKKPGSSVKVSCKASGYSFTLGYIH WVRQAPGQGLEWMGWFFPGSGNIKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.53 SEQ ID NO: 222 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFFPGSGNIKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVGINVVWHQ QKPGKAPKALIYSSSHRYSGVPSRFSGSGSGTEFTLTI SSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 223 VH QVQLVQSGAEVKKPGSSVKVSCKASGYSFRLTYIH WVRQAPGQGLEWMGWFFPGSGNIKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.54 SEQ ID NO: 224 VH + VL QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFFPGSGNIKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVGINVVWHQ QKPGKAPKALIYSSSHRYSGVPSRFSGSGSGTEFTLTI SSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: 225 VH QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH WVRQAPGQGLEWMGWFFPGSGNIKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS A-H.55 antibody SEQ ID NO: 3 HC CDR1 (Combined) GYSFTTYYIH SEQ ID NO: 4 HC CDR2 (Combined) WFFPGSGNIKYNEKFKG SEQ ID NO: 5 HC CDR3 (Combined) SYYSYDVLDY SEQ ID NO: 45 HC CDR1 (Kabat) TYYIH SEQ ID NO: 46 HC CDR2 (Kabat) WFFPGSGNIKYNEKFKG SEQ ID NO: 47 HC CDR3 (Kabat) SYYSYDVLDY SEQ ID NO: 48 HC CDR1 (Chothia) GYSFTTY SEQ ID NO: 49 HC CDR2 (Chothia) FPGSGN SEQ ID NO: 50 HC CDR3 (Chothia) SYYSYDVLDY SEQ ID NO: VH QVQLVQSGAEVKKPGSSVKVSCKASGYSFTTYYIH 1100 WVRQAPGQGLEWMGWFFPGSGNIKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS SEQ ID NO: 6 LC CDR1 (Combined) KASQNVGINVV SEQ ID NO: 7 LC CDR2 (Combined) SSSHRYS SEQ ID NO: 8 LC CDR3 (Combined) QQFKSYPLT SEQ ID NO: 51 LC CDR1 (Kabat) KASQNVGINVV SEQ ID NO: 52 LC CDR2 (Kabat) SSSHRYS SEQ ID NO: 53 LC CDR3 (Kabat) QQFKSYPLT SEQ ID NO: 54 LC CDR1 (Chothia) KASQNVGINVV SEQ ID NO: 55 LC CDR2 (chothia) SSSHRYS SEQ ID NO: 56 LC CDR3 (chothia) QQFKSYPLT SEQ ID NO: VL QSVLTQPPSVSEAPRQRVTISCKASQNVGINVVWHQ 1101 QLPGKAPKALIYSSSHRYSGVSDRFSGSGSGTSFSLAI SGLQSEDEADYFCQQFKSYPLTFGTGTKVTVL A-H.56 SEQ ID NO: VH + VL (ScFv) QVQLVQSGAEVKKPGSSVKVSCKASGHDFDKFYIH 1309 WVRQAPGQGLEWMGRVSAGSGNVKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYD VLDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGG SDIQMTQSPSFLSASVGDRVTITCKASQNVGNRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK A-H.57 SEQ ID NO: VH + VL (ScFv) QVQLVQSGAEVKKPGSSVKVSCKASGHDFRLTYIH 1326 WVRQAPGQGLEWMGRVSAGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVGDRVVW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK A-H.58 SEQ ID NO: VH + VL (ScFv) QVQLVQSGAEVKKPGSSVKVSCKASGHDFRLTYIH 1327 WVRQAPGQGLEWMGRVSAGSGNVKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYD VLDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGG SDIQMTQSPSFLSASVGDRVTITCKASQNVGNRVVW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK A-H.59 SEQ ID NO: VH + VL (ScFv) QVQLVQSGAEVKKPGSSVKVSCKASGHDFRLTYIH 1328 WVRQAPGQGLEWMGRIYAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVADRVVW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK A-H.60 SEQ ID NO: VH + VL (ScFv) QVQLVQSGAEVKKPGSSVKVSCKASGHDFKLTYIH 1329 WVRQAPGQGLEWMGRVSAGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVGDRVAW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK A-H.61 SEQ ID NO: VH + VL (ScFv) QVQLVQSGAEVKKPGSSVKVSCKASGHDFKLTYIH 1330 WVRQAPGQGLEWMGRVSAGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVDNRVAW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK A-H.62 SEQ ID NO: VH + VL (ScFv) QVQLVQSGAEVKKPGSSVKVSCKASGHDFRLTYIH 1331 WVRQAPGQGLEWMGRVSAGSGNVKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYD VLDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGG SDIQMTQSPSFLSASVGDRVTITCKASQNVADRVVW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK A-H.63 SEQ ID NO: VH + VL (ScFv) QVQLVQSGAEVKKPGSSVKVSCKASGHDFRLTYIH 1332 WVRQAPGQGLEWMGRVYAGSGNTKYNEKFKGRV TITADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYD VLDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGG SDIQMTQSPSFLSASVGDRVTITCKASQNVEDRVVW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK A-H.64 SEQ ID NO: VH + VL (ScFv) QVQLVQSGAEVKKPGSSVKVSCKASGHDFKLTYIH 1333 WVRQAPGQGLEWMGRVSAGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVADRVVW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK A-H.65 SEQ ID NO: VH + VL (ScFv) QVQLVQSGAEVKKPGSSVKVSCKASGHDFKLTYIH 1334 WVRQAPGQGLEWMGRISAGSGNTKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVGDRVVWHQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK A-H.66 SEQ ID NO: VH + VL (ScFv) QVQLVQSGAEVKKPGSSVKVSCKASGHDFKLTYIH 1335 WVRQAPGQGLEWMGRIYAGSGNTKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVGDRVVW HQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK A-H.67 SEQ ID NO: VH + VL (ScFv) QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH 1336 WVRQAPGQGLEWMGRIFPGSGNVKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVDNRVAWYQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK A-H.68 SEQ ID NO: VH + VL (ScFv) QVQLVQSGAEVKKPGSSVKVSCKASGHDFRLTYIH 1337 WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYDV LDYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DIQMTQSPSFLSASVGDRVTITCKASQNVADRVAW YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK A-H.69 (also referred to as A-H.13) SEQ ID NO: 110 VH + VL (ScFv) QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH WVRQAPGQGLEWMGRIFPGSGNVKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSDI QMTQSPSFLSASVGDRVTITCKASQNVDNRVAWYQ QKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFTLT ISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK A-H humanized-matured VH SEQ ID NO: VH-humanized matured QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH 1310 1 WVRQAPGQGLEWMGRIFPGSGNVKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAGSYYSYDVL DYWGQGTTVTVSS SEQ ID NO: VH-humanized matured QVQLVQSGAEVKKPGSSVKVSCKASGTDFKLTYIH 1311 2 WVRQAPGQGLEWMGRIFPGSGNVKYNEKFKGRVTI TADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYDVL DYWGQGTTVTVSS SEQ ID NO: VH-humanized matured QVQLVQSGAEVKKPGSSVKVSCKASGHDFRLTYIH 1312 3 WVRQAPGQGLEWMGRISAGSGNVKYNEKFKGRVT ITADTSTSTAYMELSSLRSEDTAVYYCAVSYYSYDV LDYWGQGTTVTVSS A-H humanized-matured VL SEQ ID NO: VL-humanized matured DIQMTQSPSFLSASVGDRVTITCKASQNVDNRVAW 1313 1 YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK SEQ ID NO: VL-humanized matured DIQMTQSPSFLSASVGDRVTITCKASQNVADRVAW 1314 2 YQQKPGKAPKALIYSSSHRYKGVPSRFSGSGSGTEFT LTISSLQPEDFATYFCQQFKSYPLTFGQGTKLEIK A-H.70 SEQ ID NO: 1346 VH QVQLVQSGAEVKKPGSSVK (CDRs underlined) VSCKASGHDFRLTYIHWVR QAPGQGLEWMGRVSAGSGN VKYNEKFKGRVTITADTSTS TAYMELSSLRSEDTAVYYCA VSYYSYDVLDYWGQGTTVT VSS SEQ ID NO: 1347 VL DIQMTQSPSFLSASVGDRVTI (CDRs underlined) TCKASQNVGNRVVWHQQR PGKAPKALIYSSSHRYKGVP SRFSGSGSGTEFTLTISSLQPE DFATYFCQQFKSYPLTFGQG TKLEIK A-H.71 SEQ ID NO: 1348 VH QVQLVQSGAEVKKPGSSVK (CDRs underlined) VSCKASGHDFRLTYIHWVR QAPGQGLEWMGRIYAGSGN VKYNEKFKGRVTITADTSTS TAYMELSSLRSEDTAVYYCA VSYYSYDVLDYWGQGTTVT VSS SEQ ID NO: 1349 VL DIQMTQSPSFLSASVGDRVTI (CDRs underlined) TCKASQNVADRVVWHQQK PGKAPKALIYSSSHRYKGVP SRFSGSGSGTEFTLTISSLQPE DFATYFCQQFRSYPLTFGQG TKLEIK A-H.72 SEQ ID NO: 1350 VH QVQLVQSGAEVKKPGSSVK (CDRs underlined) VSCKASGHDFRLTYIHWVR QAPGQGLEWMGRVSAGSGN TKYNEKFKGRVTITADTSTS TAYMELSSLRSEDTAVYYCA VSYYSYDVLDYWGQGTTVT VSS SEQ ID NO: 1351 VL DIQMTQSPSFLSASVGDRVTI (CDRs underlined) TCKASQNVGDRVAWHQQK PGKAPKALIYSSSHRYKGVP SRFSGSGSGTEFTLTISSLQPE DFATYFCQQFRSYPLTFGQG TKLEIK A-H.73 SEQ ID NO: 1350 VH QVQLVQSGAEVKKPGSSVK (CDRs underlined) VSCKASGHDFRLTYIHWVR QAPGQGLEWMGRVSAGSGN TKYNEKFKGRVTITADTSTS TAYMELSSLRSEDTAVYYCA VSYYSYDVLDYWGQGTTVT VSS SEQ ID NO: 1353 VL DIQMTQSPSFLSASVGDRVTI (CDRs underlined) TCKASQNVDNRVAWHQQK PGKAPKALIYSSSHRYKGVP SRFSGSGSGTEFTLTISSLQPE DFATYFCQQFKSYPLTFGQG TKLEIK A-H.74 SEQ ID NO: 1346 VH QVQLVQSGAEVKKPGSSVK (CDRs underlined) VSCKASGHDFRLTYIHWVR QAPGQGLEWMGRVSAGSGN VKYNEKFKGRVTITADTSTS TAYMELSSLRSEDTAVYYCA VSYYSYDVLDYWGQGTTVT VSS SEQ ID NO: 1349 VL DIQMTQSPSFLSASVGDRVTI (CDRs underlined) TCKASQNVADRVVWHQQK PGKAPKALIYSSSHRYKGVP SRFSGSGSGTEFTLTISSLQPE DFATYFCQQFKSYPLTFGQG TKLEIK A-H.75 SEQ ID NO: 1356 VH QVQLVQSGAEVKKPGSSVK (CDRs underlined) VSCKASGHDFRLTYIHWVR QAPGQGLEWMGRVYAGSG NTKYNEKFKGRVTITADTST STAYMELSSLRSEDTAVYYC AVSYYSYDVLDYWGQGTTV TVSS SEQ ID NO: 1357 VL DIQMTQSPSFLSASVGDRVTI (CDRs underlined) TCKASQNVEDRVVWHQQKP GKAPKALIYSSSHRYKGVPS RFSGSGSGTEFTLTISSLQPED FATYFCQQFKSYPLTFGQGT KLEIK A-H.76 SEQ ID NO: 1350 VH QVQLVQSGAEVKKPGSSVK (CDRs underlined) VSCKASGHDFRLTYIHWVR QAPGQGLEWMGRVSAGSGN TKYNEKFKGRVTITADTSTS TAYMELSSLRSEDTAVYYCA VSYYSYDVLDYWGQGTTVT VSS SEQ ID NO: 1349 VL DIQMTQSPSFLSASVGDRVTI (CDRs underlined) TCKASQNVADRVVWHQQK PGKAPKALIYSSSHRYKGVP SRFSGSGSGTEFTLTISSLQPE DFATYFCQQFKSYPLTFGQG TKLEIK A-H.77 SEQ ID NO: 1360 VH QVQLVQSGAEVKKPGSSVK (CDRs underlined) VSCKASGHDFKLTYIHWVR QAPGQGLEWMGRISAGSGN TKYNEKFKGRVTITADTSTS TAYMELSSLRSEDTAVYYCA VSYYSYDVLDYWGQGTTVT VSS SEQ ID NO: 1361 VL DIQMTQSPSFLSASVGDRVTI (CDRs underlined) TCKASQNVGDRVVWHQQK PGKAPKALIYSSSHRYKGVP SRFSGSGSGTEFTLTISSLQPE DFATYFCQQFKSYPLTFGQG TKLEIK A-H.78 SEQ ID NO: 1362 VH QVQLVQSGAEVKKPGSSVK (CDRs underlined) VSCKASGHDFKLTYIHWVR QAPGQGLEWMGRIYAGSGN TKYNEKFKGRVTITADTSTS TAYMELSSLRSEDTAVYYCA VSYYSYDVLDYWGQGTTVT VSS SEQ ID NO: 1361 VL DIQMTQSPSFLSASVGDRVTI (CDRs underlined) TCKASQNVGDRVVWHQQK PGKAPKALIYSSSHRYKGVP SRFSGSGSGTEFTLTISSLQPE DFATYFCQQFKSYPLTFGQG TKLEIK A-H.79 SEQ ID NO: 1350 VH QVQLVQSGAEVKKPGSSVK (CDRs underlined) VSCKASGHDFKLTYIHWVR QAPGQGLEWMGRVSAGSGN TKYNEKFKGRVTITADTSTS TAYMELSSLRSEDTAVYYCA VSYYSYDVLDYWGQGTTVT VSS SEQ ID NO: 1365 VL DIQMTQSPSFLSASVGDRVTI (CDRs underlined) TCRASQNVDNRLGWHQQKP GKAPKALIYSSSHRYKGVPS RFSGSGSGTEFTLTISSLQPED FATYFCQQFKSYPLTFGQGT KLEIK A-H.80 SEQ ID NO: 1350 VH QVQLVQSGAEVKKPGSSVK (CDRs underlined) VSCKASGHDFKLTYIHWVR QAPGQGLEWMGRVSAGSGN TKYNEKFKGRVTITADTSTS TAYMELSSLRSEDTAVYYCA VSYYSYDVLDYWGQGTTVT VSS SEQ ID NO: 1367 VL DIQMTQSPSFLSASVGDRVTI (CDRs underlined) TCKASQNVDNRVAWHQQK PGKAPKALIYAASSLQKGVP SRFSGSGSGTEFTLTISSLQPE DFATYFCQQFKSYPLTFGQG TKLEIK A-H.81 SEQ ID NO: 1350 VH QVQLVQSGAEVKKPGSSVK (CDRs underlined) VSCKASGHDFKLTYIHWVR QAPGQGLEWMGRVSAGSGN TKYNEKFKGRVTITADTSTS TAYMELSSLRSEDTAVYYCA VSYYSYDVLDYWGQGTTVT VSS SEQ ID NO: 1369 VL DIQMTQSPSFLSASVGDRVTI (CDRs underlined) TCKASQNVDNRVAWHQQK PGKAPKALIYSSSHRYKGVP SRFSGSGSGTEFTLTISSLQPE DFATYFCLQHNSYPLTFGQG TKLEIK A-H.82 SEQ ID NO: 1370 VH QVQLVQSGAEVKKPGSSVK (CDRs underlined) VSCKASGHDFKLTYIHWVR QAPGQGLEWMGRVSAGSGN VNYAQKFQGRVTITADTSTS TAYMELSSLRSEDTAVYYCA VSYYSYDVLDYWGQGTTVT VSS SEQ ID NO: 1365 VL DIQMTQSPSFLSASVGDRVTI (CDRs underlined) TCRASQNVDNRLGWHQQKP GKAPKALIYSSSHRYKGVPS RFSGSGSGTEFTLTISSLQPED FATYFCQQFKSYPLTFGQGT KLEIK A-H.83 SEQ ID NO: 1370 VH QVQLVQSGAEVKKPGSSVK (CDRs underlined) VSCKASGHDFKLTYIHWVR QAPGQGLEWMGRVSAGSGN VNYAQKFQGRVTITADTSTS TAYMELSSLRSEDTAVYYCA VSYYSYDVLDYWGQGTTVT VSS SEQ ID NO: 1367 VL DIQMTQSPSFLSASVGDRVTI (CDRs underlined) TCKASQNVDNRVAWHQQK PGKAPKALIYAASSLQKGVP SRFSGSGSGTEFTLTISSLQPE DFATYFCQQFKSYPLTFGQG TKLEIK A-H.84 SEQ ID NO: 1370 VH QVQLVQSGAEVKKPGSSVK (CDRs underlined) VSCKASGHDFKLTYIHWVR QAPGQGLEWMGRVSAGSGN VNYAQKFQGRVTITADTSTS TAYMELSSLRSEDTAVYYCA VSYYSYDVLDYWGQGTTVT VSS SEQ ID NO: 1369 VL DIQMTQSPSFLSASVGDRVTI (CDRs underlined) TCKASQNVDNRVAWHQQK PGKAPKALIYSSSHRYKGVP SRFSGSGSGTEFTLTISSLQPE DFATYFCLQHNSYPLTFGQG TKLEIK A-H.85 SEQ ID NO: 1344 VH (CDRs underlined) QVQLVQSGAEVKKPGSSVK VSCKASGHDFRLTYIHWVR QAPGQGLEWMGRVSAGSGN TKYNEKFKGRVTITADTSTS TAYMELSSLRSEDTAVYYCA VSYYSYDVLDYWGQGTTVT VSS SEQ ID NO: 1361 VL DIQMTQSPSFLSASVGDRVTI (CDRs underlined) TCKASQNVGDRVVWHQQK PGKAPKALIYSSSHRYKGVP SRFSGSGSGTEFTLTISSLQPE DFATYFCQQFKSYPLTFGQG TKLEIK

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule comprises a VH and/or a VL of an antibody described in Table 1, or a sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule comprises a VH and a VL of an antibody described in Table 1, or a sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.

In some embodiments, an anti-TCRVb antibody disclosed herein has an antigen binding domain having a VL having a consensus sequence of SEQ ID NO: 230, wherein position 30 is G, E, A or D; position 31 is N or D; position 32 is R or K; position 36 is Y or H; and/or position 56 is K or S.

In some embodiments, an anti-TCRVb antibody disclosed herein has an antigen binding domain having a VH having a consensus sequence of SEQ ID NO: 231, wherein: position 27 is H or T or G or Y; position 28 is D or T or S; position 30 is H or R or D or K or T; position 31 is L or D or K or T or N; position 32 is W or F or T or I or Y or G; position 49 is R or W; position 50 is V or I or F; position 51 is F or S or Y; position 52 is A or P; position 56 is N or S; position 57 is T or V or Y or I; position 58 is K or R; position 97 is G or V; position 99 is Y or I; position 102 is Y or A; and/or position 103 is D or G.

Anti-TCRβ V12 Antibodies

Accordingly, in one aspect, the disclosure provides an anti-TCRβV antibody molecule that binds to human TCRβ V12, e.g., a TCRβ V12 subfamily comprising: TCRβ V12-4*01, TCRβ V12-3*01 or TCRβ V12-5*01. In some embodiments the TCRβ V12 subfamily comprises TCRβ V12-4*01. In some embodiments the TCRβ V12 subfamily comprises TCRβ V12-3*01.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, is a non-murine antibody molecule, e.g., a human or humanized antibody molecule. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule is a human antibody molecule. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule is a humanized antibody molecule.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, is isolated or recombinant.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, comprises at least one antigen-binding region, e.g., a variable region or an antigen-binding fragment thereof, from an antibody described herein, e.g., an antibody described in Table 2, or encoded by a nucleotide sequence in Table 2, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, comprises at least one, two, three or four variable regions from an antibody described herein, e.g., an antibody as described in Table 2, or encoded by a nucleotide sequence in Table 2, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, comprises at least one or two heavy chain variable regions from an antibody described herein, e.g., an antibody as described in Table 2, or encoded by a nucleotide sequence in Table 2, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, comprises at least one or two light chain variable regions from an antibody described herein, e.g., an antibody as described in Table 2, or encoded by a nucleotide sequence in Table 2, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, comprises a heavy chain constant region for an IgG4, e.g., a human IgG4. In still another embodiment, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, includes a heavy chain constant region for an IgG1, e.g., a human IgG1. In one embodiment, the heavy chain constant region comprises an amino sequence set forth in Table 3, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) thereto.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, includes a kappa light chain constant region, e.g., a human kappa light chain constant region. In one embodiment, the light chain constant region comprises an amino sequence set forth in Table 3, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) thereto.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, includes at least one, two, or three complementarity determining regions (CDRs) from a heavy chain variable region of an antibody described herein, e.g., an antibody as described in Table 2, or encoded by the nucleotide sequence in Table 2, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, includes at least one, two, or three CDRs (or collectively all of the CDRs) from a heavy chain variable region comprising an amino acid sequence shown in Table 2, or encoded by a nucleotide sequence shown in Table 2. In one embodiment, one or more of the CDRs (or collectively all of the CDRs) have one, two, three, four, five, six or more changes, e.g., amino acid substitutions or deletions, relative to the amino acid sequence shown in Table 2, or encoded by a nucleotide sequence shown in Table 2.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, includes at least one, two, or three complementarity determining regions (CDRs) from a light chain variable region of an antibody described herein, e.g., an antibody as described in Table 2, or encoded by the nucleotide sequence in Table 2, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, includes at least one, two, or three CDRs (or collectively all of the CDRs) from a light chain variable region comprising an amino acid sequence shown in Table 2, or encoded by a nucleotide sequence shown in Table 2. In one embodiment, one or more of the CDRs (or collectively all of the CDRs) have one, two, three, four, five, six or more changes, e.g., amino acid substitutions or deletions, relative to the amino acid sequence shown in Table 2, or encoded by a nucleotide sequence shown in Table 2.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, includes at least one, two, three, four, five or six CDRs (or collectively all of the CDRs) from a heavy and light chain variable region comprising an amino acid sequence shown in Table 2, or encoded by a nucleotide sequence shown in Table 2. In one embodiment, one or more of the CDRs (or collectively all of the CDRs) have one, two, three, four, five, six or more changes, e.g., amino acid substitutions or deletions, relative to the amino acid sequence shown in Table 2, or encoded by a nucleotide sequence shown in Table 2.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, molecule includes all six CDRs from an antibody described herein, e.g., an antibody as described in Table 2, or encoded by the nucleotide sequence in Table 2, or closely related CDRs, e.g., CDRs which are identical or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions). In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, may include any CDR described herein.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes at least one, two, or three CDRs according to Kabat et al. (e.g., at least one, two, or three CDRs according to the Kabat definition as set out in Table 2) from a heavy chain variable region of an antibody described herein, e.g., an antibody chosen as described in Table 2, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to one, two, or three CDRs according to Kabat et al. shown in Table 2.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes at least one, two, or three CDRs according to Kabat et al. (e.g., at least one, two, or three CDRs according to the Kabat definition as set out in Table 2) from a light chain variable region of an antibody described herein, e.g., an antibody as described in Table 2, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to one, two, or three CDRs according to Kabat et al. shown in Table 2.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes at least one, two, three, four, five, or six CDRs according to Kabat et al. (e.g., at least one, two, three, four, five, or six CDRs according to the Kabat definition as set out in Table 2) from the heavy and light chain variable regions of an antibody described herein, e.g., an antibody as described in Table 2, or encoded by the nucleotide sequence in Table 2; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to one, two, three, four, five, or six CDRs according to Kabat et al. shown in Table 2.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes all six CDRs according to Kabat et al. (e.g., all six CDRs according to the Kabat definition as set out in Table 2) from the heavy and light chain variable regions of an antibody described herein, e.g., an antibody as described in Table 2, or encoded by the nucleotide sequence in Table 2; or encoded by the nucleotide sequence in Table 2; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to all six CDRs according to Kabat et al. shown in Table 2. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule may include any CDR described herein.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes at least one, two, or three hypervariable loops that have the same canonical structures as the corresponding hypervariable loop of an antibody described herein, e.g., an antibody described in Table 2, e.g., the same canonical structures as at least loop 1 and/or loop 2 of the heavy and/or light chain variable domains of an antibody described herein. See, e.g., Chothia et al., (1992) J. Mol. Biol. 227:799-817; Tomlinson et al., (1992) J. Mol. Biol. 227:776-798 for descriptions of hypervariable loop canonical structures. These structures can be determined by inspection of the tables described in these references.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes at least one, two, or three CDRs according to Chothia et al. (e.g., at least one, two, or three CDRs according to the Chothia definition as set out in Table 2) from a heavy chain variable region of an antibody described herein, e.g., an antibody chosen as described in Table 2, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to one, two, or three CDRs according to Chothia et al. shown in Table 2.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes at least one, two, or three CDRs according to Chothia et al. (e.g., at least one, two, or three CDRs according to the Chothia definition as set out in Table 2) from a light chain variable region of an antibody described herein, e.g., an antibody as described in Table 2, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to one, two, or three CDRs according to Chothia et al. shown in Table 2.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes at least one, two, three, four, five, or six CDRs according to Chothia et al. (e.g., at least one, two, three, four, five, or six CDRs according to the Chothia definition as set out in Table 2) from the heavy and light chain variable regions of an antibody described herein, e.g., an antibody as described in Table 2, or encoded by the nucleotide sequence in Table 2; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to one, two, three, four, five, or six CDRs according to Chothia et al. shown in Table 2.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes all six CDRs according to Chothia et al. (e.g., all six CDRs according to the Chothia definition as set out in Table 2) from the heavy and light chain variable regions of an antibody described herein, e.g., an antibody as described in Table 2, or encoded by the nucleotide sequence in Table 2; or encoded by the nucleotide sequence in Table 2; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to all six CDRs according to Chothia et al. shown in Table 2. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule may include any CDR described herein.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes at least one, two, or three CDRs according to a combined CDR (e.g., at least one, two, or three CDRs according to the combined CDR definition as set out in Table 2) from a heavy chain variable region of an antibody described herein, e.g., an antibody chosen as described in Table 2, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to one, two, or three CDRs according to combined CDR shown in Table 2.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes at least one, two, or three CDRs according to a combined CDR (e.g., at least one, two, or three CDRs according to the combined CDR definition as set out in Table 2) from a light chain variable region of an antibody described herein, e.g., an antibody as described in Table 2, or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to one, two, or three CDRs according to a combined CDR shown in Table 2.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes at least one, two, three, four, five, or six CDRs according to a combined CDR. (e.g., at least one, two, three, four, five, or six CDRs according to the combined CDR definition as set out in Table 2) from the heavy and light chain variable regions of an antibody described herein, e.g., an antibody as described in Table 2, or encoded by the nucleotide sequence in Table 2; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to one, two, three, four, five, or six CDRs according to a combined CDR shown in Table 2.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes all six CDRs according to a combined CDR (e.g., all six CDRs according to the combined CDR definition as set out in Table 2) from the heavy and light chain variable regions of an antibody described herein, e.g., an antibody as described in Table 2, or encoded by the nucleotide sequence in Table 2; or encoded by the nucleotide sequence in Table 2; or a sequence substantially identical (e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical) to any of the aforesaid sequences; or which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) relative to all six CDRs according to a combined CDR shown in Table 2. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule may include any CDR described herein.

In some embodiments, a combined CDR as set out in Table 1 is a CDR that comprises a Kabat CDR and a Chothia CDR.

In some embodiments, the anti-TCRβV antibody molecule, e e.g., anti-TCRβ V12 antibody molecule, molecule includes a combination of CDRs or hypervariable loops identified as combined CDRs in Table 1. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, can contain any combination of CDRs or hypervariable loops according the “combined” CDRs are described in Table 1.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes a combination of CDRs or hypervariable loops defined according to the Kabat et al. and Chothia et al., or as described in Table 1.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule can contain any combination of CDRs or hypervariable loops according to the Kabat and Chothia definitions.

In an embodiment, e.g., an embodiment comprising a variable region, a CDR (e.g., a combined CDR, Chothia CDR or Kabat CDR), or other sequence referred to herein, e.g., in Table 2, the antibody molecule is a monospecific antibody molecule, a bispecific antibody molecule, a bivalent antibody molecule, a biparatopic antibody molecule, or an antibody molecule that comprises an antigen binding fragment of an antibody, e.g., a half antibody or antigen binding fragment of a half antibody. In certain embodiments the antibody molecule comprises a multispecific molecule, e.g., a bispecific molecule, e.g., as described herein.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes:

    • (i) one, two or all of a light chain complementarity determining region 1 (LC CDR1), a light chain complementarity determining region 2 (LC CDR2), and a light chain complementarity determining region 3 (LC CDR3) of SEQ ID NO: 16, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO: 30, and/or
    • (ii) one, two or all of a heavy chain complementarity determining region 1 (HC CDR1), heavy chain complementarity determining region 2 (HC CDR2), and a heavy chain complementarity determining region 3 (HC CDR3) of SEQ ID NO: 15, SEQ ID NO: 23, SEQ ID NO: 24 or SEQ ID NO: 25.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • (i) a LC CDR1 amino acid sequence of SEQ ID NO: 20, a LC CDR2 amino acid sequence of SEQ ID NO: 21, or a LC CDR3 amino acid sequence of SEQ ID NO: 22; and/or
    • (ii) a HC CDR1 amino acid sequence of SEQ ID NO: 17, a HC CDR2 amino acid sequence of SEQ ID NO: 18, or a HC CDR3 amino acid sequence of SEQ ID NO: 19.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • (i) a light chain variable region (VL) comprising a LC CDR1 amino acid sequence of SEQ ID NO: 20, a LC CDR2 amino acid sequence of SEQ ID NO: 21, and a LC CDR3 amino acid sequence of SEQ ID NO: 2; and/or
    • (ii) a heavy chain variable region (VH) comprising a HC CDR1 amino acid sequence of SEQ ID NO: 17, a HC CDR2 amino acid sequence of SEQ ID NO: 18, and a HC CDR3 amino acid sequence of SEQ ID NO: 19.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • (i) a LC CDR1 amino acid sequence of SEQ ID NO: 63, a LC CDR2 amino acid sequence of SEQ ID NO: 64, or a LC CDR3 amino acid sequence of SEQ ID NO: 65; and/or
    • (ii) a HC CDR1 amino acid sequence of SEQ ID NO: 57, a HC CDR2 amino acid sequence of SEQ ID NO: 58, or a HC CDR3 amino acid sequence of SEQ ID NO: 59.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • (i) a light chain variable region (VL) comprising a LC CDR1 amino acid sequence of SEQ ID NO: 63, a LC CDR2 amino acid sequence of SEQ ID NO: 64, or a LC CDR3 amino acid sequence of SEQ ID NO: 65; and/or
    • (ii) a heavy chain variable region (VH) comprising a HC CDR1 amino acid sequence of SEQ ID NO: 57, a HC CDR2 amino acid sequence of SEQ ID NO: 58, or a HC CDR3 amino acid sequence of SEQ ID NO: 59.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • (i) a LC CDR1 amino acid sequence of SEQ ID NO: 66, a LC CDR2 amino acid sequence of SEQ ID NO: 67, or a LC CDR3 amino acid sequence of SEQ ID NO: 68; and/or
    • (ii) a HC CDR1 amino acid sequence of SEQ ID NO: 60, a HC CDR2 amino acid sequence of SEQ ID NO: 61, or a HC CDR3 amino acid sequence of SEQ ID NO: 62.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • (i) a light chain variable region (VL) comprising a LC CDR1 amino acid sequence of SEQ ID NO: 63, a LC CDR2 amino acid sequence of SEQ ID NO: 64, or a LC CDR3 amino acid sequence of SEQ ID NO: 65; and/or
    • (ii) a heavy chain variable region (VH) comprising a HC CDR1 amino acid sequence of SEQ ID NO: 57, a HC CDR2 amino acid sequence of SEQ ID NO: 58, or a HC CDR3 amino acid sequence of SEQ ID NO: 59.

In one embodiment, the light or the heavy chain variable framework (e.g., the region encompassing at least FR1, FR2, FR3, and optionally FR4) of the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule can be chosen from: (a) a light or heavy chain variable framework including at least 80%, 85%, 87% 90%, 92%, 93%, 95%, 97%, 98%, or 100% of the amino acid residues from a human light or heavy chain variable framework, e.g., a light or heavy chain variable framework residue from a human mature antibody, a human germline sequence, or a human consensus sequence; (b) a light or heavy chain variable framework including from 20% to 80%, 40% to 60%, 60% to 90%, or 70% to 95% of the amino acid residues from a human light or heavy chain variable framework, e.g., a light or heavy chain variable framework residue from a human mature antibody, a human germline sequence, or a human consensus sequence; (c) a non-human framework (e.g., a rodent framework); or (d) a non-human framework that has been modified, e.g., to remove antigenic or cytotoxic determinants, e.g., deimmunized, or partially humanized. In one embodiment, the light or heavy chain variable framework region (particularly FR1, FR2 and/or FR3) includes a light or heavy chain variable framework sequence at least 70, 75, 80, 85, 87, 88, 90, 92, 94, 95, 96, 97, 98, 99% identical or identical to the frameworks of a VL or VH segment of a human germline gene.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, comprises a heavy chain variable domain having at least one, two, three, four, five, six, seven, ten, fifteen, twenty or more changes, e.g., amino acid substitutions or deletions, from an amino acid sequence described in Table 2.e.g., the amino acid sequence of the FR region in the entire variable region, e.g., shown in FIGS. 2A and 2B, or in SEQ ID NOs: 23-25.

Alternatively, or in combination with the heavy chain substitutions described herein the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain variable domain having at least one, two, three, four, five, six, seven, ten, fifteen, twenty or more amino acid changes, e.g., amino acid substitutions or deletions, from an amino acid sequence of an antibody described herein e.g., the amino acid sequence of the FR region in the entire variable region, e.g., shown in FIGS. 2A and 2B, or in SEQ ID NOs: 26-30.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes one, two, three, or four heavy chain framework regions shown in FIG. 2A, or a sequence substantially identical thereto.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes one, two, three, or four light chain framework regions shown in FIG. 2B, or a sequence substantially identical thereto.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises the light chain framework region 1 e.g., as shown in FIG. 2B.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises the light chain framework region 2 e.g., as shown in FIG. 2B.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises the light chain framework region 3, e.g., as shown in FIG. 2B.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises the light chain framework region 4, e.g., as shown in FIG. 2B.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain comprising a framework region, e.g., framework region 1 (FR1), comprising a change, e.g., a substitution (e.g., a conservative substitution) at one or more, e.g., all, position disclosed herein according to Kabat numbering. In some embodiments, FR1 comprises an Aspartic Acid at position 1, e.g., a substitution at position 1 according to Kabat numbering, e.g., an Alanine to Aspartic Acid substitution. In some embodiments, FR1 comprises an Asparagine at position 2, e.g., a substitution at position 2 according to Kabat numbering, e.g., an Isoleucine to Asparagine substitution, Serine to Asparagine substitution or Tyrosine to Asparagine substitution. In some embodiments, FR1 comprises a Leucine at position 4, e.g., a substitution at position 4 according to Kabat numbering, e.g., a Methionine to Leucine substitution.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain comprising a framework region, e.g., framework region 1 (FR1), comprising a substitution at position 1 according to Kabat numbering, e.g., an Alanine to Aspartic Acid substitution, a substitution at position 2 according to Kabat numbering, e.g., an Isoleucine to Asparagine substitution, Serine to Asparagine substitution or Tyrosine to Asparagine substitution, and a substitution at position 4 according to Kabat numbering, e.g., a Methionine to Leucine substitution. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain comprising a framework region, e.g., framework region 1 (FR1), comprising a substitution at position 1 according to Kabat numbering, e.g., an Alanine to Aspartic Acid substitution, and a substitution at position 2 according to Kabat numbering, e.g., an Isoleucine to Asparagine substitution, Serine to Asparagine substitution or Tyrosine to Asparagine substitution. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain comprising a framework region, e.g., framework region 1 (FR1), comprising a substitution at position 1 according to Kabat numbering, e.g., an Alanine to Aspartic Acid substitution, and a substitution at position 4 according to Kabat numbering, e.g., a Methionine to Leucine substitution. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain comprising a framework region, e.g., framework region 1 (FR1), comprising a substitution at position 2 according to Kabat numbering, e.g., an Isoleucine to Asparagine substitution, Serine to Asparagine substitution or Tyrosine to Asparagine substitution, and a substitution at position 4 according to Kabat numbering, e.g., a Methionine to Leucine substitution. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain comprising a framework region, e.g., framework region 3 (FR3), comprising a change, e.g., a substitution (e.g., a conservative substitution) at one or more, e.g., all, position disclosed herein according to Kabat numbering. In some embodiments, FR3 comprises a Glycine at position 66, e.g., a substitution at position 66 according to Kabat numbering, e.g., a Lysine to Glycine substitution, or a Serine to Glycine substitution. In some embodiments, FR3 comprises an Asparagine at position 69, e.g., a substitution at position 69 according to Kabat numbering, e.g., a Tyrosine to Asparagine substitution. In some embodiments, FR3 comprises a Tyrosine at position 71, e.g., a substitution at position 71 according to Kabat numbering, e.g., a Phenylalanine to Tyrosine substitution, or an Alanine to Tyrosine substitution.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain comprising a framework region, e.g., framework region 3 (FR3), comprising a substitution at position 66 according to Kabat numbering, e.g., a Lysine to Glycine substitution, or a Serine to Glycine substitution, and a substitution at position 69 according to Kabat numbering, e.g., a Tyrosine to Asparagine substitution. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain comprising a framework region, e.g., framework region 3 (FR3), comprising a substitution at position 66 according to Kabat numbering, e.g., Lysine to Glycine substitution, or a Serine to Glycine substitution, and a substitution at position 71 according to Kabat numbering, e.g., a Phenylalanine to Tyrosine substitution, or an Alanine to Tyrosine substitution. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain comprising a framework region, e.g., framework region 3 (FR3), comprising a substitution at position 69 according to Kabat numbering, e.g., a Tyrosine to Asparagine substitution and a substitution at position 71 according to Kabat numbering, e.g., a Phenylalanine to Tyrosine substitution, or an Alanine to Tyrosine substitution. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain comprising a framework region, e.g., framework region 3 (FR3), comprising a substitution at position 66 according to Kabat numbering, e.g., a Lysine to Glycine substitution, or a Serine to Glycine substitution, a substitution at position 69 according to Kabat numbering, e.g., a Tyrosine to Asparagine substitution and a substitution at position 71 according to Kabat numbering, e.g., a Phenylalanine to Tyrosine substitution, or an Alanine to Tyrosine substitution. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain comprising: a framework region 1 (FR1) comprising a substitution at position 2 according to Kabat numbering, e.g., a Isoleucine to Asparagine substitution; and a framework region 3 (FR3), comprising a substitution at position 69 according to Kabat numbering, e.g., a Threonine to Asparagine substitution and a substitution at position 71 according to Kabat numbering, e.g., a Phenylalanine to Tyrosine substitution, e.g., as shown in the amino acid sequence of SEQ ID NO: 26. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain comprising: (a) a framework region 1 (FR1) comprising a substitution at position 1 according to Kabat numbering, e.g., a Alanine to Aspartic Acid substitution, and a substitution at position 2 according to Kabat numbering, e.g., a Isoleucine to Asparagine substitution; and (b) a framework region 3 (FR3), comprising a substitution at position 69 according to Kabat numbering, e.g., a Threonine to Asparagine substitution and a substitution at position 71 according to Kabat numbering, e.g., a Phenylalanine to Tyrosine substitution, e.g., as shown in the amino acid sequence of SEQ ID NO: 27 In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain comprising: (a) a framework region 1 (FR1) comprising a substitution at position 2 according to Kabat numbering, e.g., a Serine to Asparagine substitution; and a substitution at position 4 according to Kabat numbering, e.g., a Methionine to Leucine substitution; and (b) a framework region 3 (FR3), comprising a substitution at position 69 according to Kabat numbering, e.g., a Threonine to Asparagine substitution and a substitution at position 71 according to Kabat numbering, e.g., a Phenylalanine to Tyrosine substitution, e.g., as shown in the amino acid sequence of SEQ ID NO: 28 In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain comprising: (a) a framework region 1 (FR1) comprising a substitution at position 2 according to Kabat numbering, e.g., a Serine to Asparagine substitution; and (b) a framework region 3 (FR3) comprising a substitution at position 66 according to Kabat numbering, e.g., a Lysine to Glycine substitution; a substitution at position 69 according to Kabat numbering, e.g., a Threonine to Asparagine substitution; and a substitution at position 71 according to Kabat numbering, e.g., a Alanine to Tyrosine substitution, e.g., as shown in the amino acid sequence of SEQ ID NO: 29. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain comprising: (a) a framework region 1 (FR1) comprising a substitution at position 2 according to Kabat numbering, e.g., a Tyrosine to Asparagine substitution; and (b) a framework region 3 (FR3) comprising a substitution at position 66 according to Kabat numbering, e.g., a Serine to Glycine substitution; a substitution at position 69 according to Kabat numbering, e.g., a Threonine to Asparagine substitution; and a substitution at position 71 according to Kabat numbering, e.g., a Alanine to Tyrosine substitution, e.g., as shown in the amino acid sequence of SEQ ID NO: 29. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises a light chain variable domain comprising: (a) a framework region 1 (FR1) comprising a change, e.g., a substitution (e.g., a conservative substitution) at one or more (e.g., all) positions disclosed herein according to Kabat numbering, and (b) a framework region 3 (FR3) comprising a change, e.g., a substitution (e.g., a conservative substitution) at one or more (e.g., all) position disclosed herein according to Kabat numbering. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises the heavy chain framework region 1, e.g., as shown in FIG. 2A.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises the heavy chain framework region 2, e.g., as shown in FIG. 2A.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises the heavy chain framework region 3, e.g., as shown in FIG. 2A.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises the heavy chain framework region 4, e.g., as shown in FIG. 2A.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises the heavy chain framework regions 1-4, e.g., SEQ ID NOS: 20-23, or as shown in FIG. 2A.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises the light chain framework regions 1-4, e.g., SEQ ID NOs: 26-30, or as shown in FIG. 2B.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises the heavy chain framework regions 1-4, e.g., SEQ ID NOs: 23-25; and the light chain framework regions 1-4, e.g., SEQ ID NOs: 26-30, or as shown in FIGS. 2A and 2B.

In some embodiments, the heavy or light chain variable domain, or both, of, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes an amino acid sequence, which is substantially identical to an amino acid disclosed herein, e.g., at least 80%, 85%, 90%, 92%, 95%, 97%, 98%, 99% or higher identical to a variable region of an antibody described herein, e.g., an antibody as described in Table 2, or encoded by the nucleotide sequence in Table 2; or which differs at least 1 or 5 residues, but less than 40, 30, 20, or 10 residues, from a variable region of an antibody described herein.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises at least one, two, three, or four antigen-binding regions, e.g., variable regions, having an amino acid sequence as set forth in Table 2, or a sequence substantially identical thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, or which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the sequences shown in Table 2. In another embodiment, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule includes a VH and/or VL domain encoded by a nucleic acid having a nucleotide sequence as set forth in Table 2, or a sequence substantially identical thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, or which differs by no more than 3, 6, 15, 30, or 45 nucleotides from the sequences shown in Table 2.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • a VH domain comprising an amino acid sequence chosen from the amino acid sequence of SEQ ID NO: 23, SEQ ID NO:24 or SEQ ID NO:25, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 23, SEQ ID NO:24 or SEQ ID NO:25, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 23, SEQ ID NO:24 or SEQ ID NO:25; and/or
    • a VL domain comprising an amino acid sequence chosen from the amino acid sequence of SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO: 30, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence of SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO: 30, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO: 30.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 23, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 23, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 23; and
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 26, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 26, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 26.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 23, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 23, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 23; and
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 27, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 27, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 27.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 23, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 23, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 23; and
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 28, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 28, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 28.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 23, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 23, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 23; and
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 29, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 29, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 29.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 23, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 23, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 23; and
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 30, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 30, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 30.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 24 or 25, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 24 or 25, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 24 or 25; and
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 26, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 26, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 26.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 24 or 25, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 24 or 25, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 24 or 25; and
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 27, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 27, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 27.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 24 or 25, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 24 or 25, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 24 or 25; and
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 28, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 28, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 28.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 24 or 25, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 24 or 25, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 24 or 25; and
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 29, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 29, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 29.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 24 or 25, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 24 or 25, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 24 or 25; and
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 30, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 30, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 30.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 25 or 23, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 25 or 23, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 25 or 23; and
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 26, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 26, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 26.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 25 or 23, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 25 or 23, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 25 or 23; and
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 27, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 27, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 27.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 25 or 23, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 25 or 23, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 25 or 23; and
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 28, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 28, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 28.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 25 or 23, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 25 or 23, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 25 or 23; and
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 29, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 29, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 29.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule comprises:

    • a VH domain comprising the amino acid sequence of SEQ ID NO: 25 or 23, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 25 or 23, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 25 or 23; and
    • a VL domain comprising the amino acid sequence of SEQ ID NO: 30, an amino acid sequence at least about 85%, 90%, 95%, 99% or more identical to the amino acid sequence SEQ ID NO: 30, or an amino acid sequence which differs by no more than 1, 2, 5, 10, or 15 amino acid residues from the amino acid sequence of SEQ ID NO: 30.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule is a full antibody or fragment thereof (e.g., a Fab, F(ab)2, Fv, or a single chain Fv fragment (scFv)). In embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V6 (e.g., anti-TCRβ V6-5*01) antibody molecule is a monoclonal antibody or an antibody with single specificity. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule, can also be a humanized, chimeric, camelid, shark, or an in vitro-generated antibody molecule. In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule is a humanized antibody molecule. The heavy and light chains of the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule can be full-length (e.g., an antibody can include at least one, and preferably two, complete heavy chains, and at least one, and preferably two, complete light chains) or can include an antigen-binding fragment (e.g., a Fab, F(ab′)2, Fv, a single chain Fv fragment, a single domain antibody, a diabody (dAb), a bivalent antibody, or bispecific antibody or fragment thereof, a single domain variant thereof, or a camelid antibody).

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule is in the form of a multispecific molecule, e.g., a bispecific molecule, e.g., as described herein.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule has a heavy chain constant region (Fc) chosen from, e.g., the heavy chain constant regions of IgG1, IgG2, IgG3, IgG4, IgM, IgA1, IgA2, IgD, and IgE. In some embodiments, the Fc region is chosen from the heavy chain constant regions of IgG1, IgG2, IgG3, and IgG4. In some embodiments, the Fc region is chosen from the heavy chain constant region of IgG1 or IgG2 (e.g., human IgG1, or IgG2). In some embodiments, the heavy chain constant region is human IgG1.

In some embodiments, the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule has a light chain constant region chosen from, e.g., the light chain constant regions of kappa or lambda, preferably kappa (e.g., human kappa). In one embodiment, the constant region is altered, e.g., mutated, to modify the properties of the anti-TCRβV antibody molecule, e.g., anti-TCRβ V12 antibody molecule (e.g., to increase or decrease one or more of: Fc receptor binding, antibody glycosylation, the number of cysteine residues, effector cell function, or complement function). For example, the constant region is mutated at positions 296 (M to Y), 298 (S to T), 300 (T to E), 477 (H to K) and 478 (N to F) to alter Fc receptor binding (e.g., the mutated positions correspond to positions 132 (M to Y), 134 (S to T), 136 (T to E), 313 (H to K) and 314 (N to F) of SEQ ID NOs: 212 or 214; or positions 135 (M to Y), 137 (S to T), 139 (T to E), 316 (H to K) and 317 (N to F) of SEQ ID NOs: 215, 216, 217 or 218).

Antibody B-H.1 comprises a first chain comprising the amino acid sequence of SEQ ID NO: 3280 and a second chain comprising the amino acid sequence of SEQ ID NO: 3281.

Additional exemplary anti-TCRβ V12 antibodies of the disclosure are provided in Table 2. In some embodiments, the anti-TCRβ V12 is antibody B, e.g., humanized antibody B (antibody B-H), as provided in Table 2. In some embodiments, the anti-TCRβV antibody comprises one or more (e.g., all three) of a LC CDR1, LC CDR2, and LC CDR3 provided in Table 2; and/or one or more (e.g., all three) of a HC CDR1, HC CDR2, and HC CDR3 provided in Table 2, or a sequence with at least 95% identity thereto. In some embodiments, antibody B comprises a variable heavy chain (VH) and/or a variable light chain (VL) provided in Table 2, or a sequence with at least 95% identity thereto.

In some embodiments, the anti-TCRVB 12 antibody molecule (e.g., anti-TCRVB 12-3 or anti-TCRVB 12-4 antibody molecule) comprises a VH of B-H.1A, B-H.1B, B-H.1C, B-H.1D, B-H.1E, B-H.1F, B-H.1G, B-H.1H, B-H.1, B-H.2, B-H.3, B-H.4, B-H.5, or B-H.6, or a sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.

In some embodiments, the anti-TCRVB 12 antibody molecule (e.g., anti-TCRVB 12-3 or anti-TCRVB 12-4 antibody molecule) comprises a VL of B-H.1A, B-H.1B, B-H.1C, B-H.1D, B-H.1E, B-H.1F, B-H.1G, B-H.1H, B-H.1, B-H.2, B-H.3, B-H.4, B-H.5, or B-H.6, or a sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.

In some embodiments, the anti-TCRVB 12 antibody molecule (e.g., anti-TCRVB 12-3 or anti-TCRVB 12-4 antibody molecule) comprises a VH of B-H.1A, B-H.1B, B-H.1C, B-H.1D, B-H.1E, B-H.1F, B-H.1G, B-H.1H, B-H.1, B-H.2, B-H.3, B-H.4, B-H.5, or B-H.6, or a sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto; and a VL of B-H.1A, B-H.1B, B-H.1C, B-H.1D, B-H.1E, B-H.1F, B-H.1G, B-H.1H, B-H.1, B-H.2, B-H.3, B-H.4, B-H.5, or B-H.6, or a sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.

TABLE 2 Amino acid and nucleotide sequences for murine and humanized antibody molecules which bind to TCRVB 12, e.g., TCRVB 12-3 or TCRVB 12-4. The antibody molecules include murine mAb Antibody B and humanized mAb Antibody B-H.1 to B-H.6. The amino acid the heavy and light chain CDRs, and the amino acid and nucleotide sequences of the heavy and light chain variable regions, and the heavy and light chains are shown. Antibody B (murine), also referred to as 16G8 SEQ ID NO: 17 HC CDR1 (Combined) GFTFSNFGMH SEQ ID NO: 18 HC CDR2 (Combined) YISSGSSTIYYADTLKG SEQ ID NO: 19 HC CDR3 (Combined) RGEGAMDY SEQ ID NO: 57 HC CDR1 (Kabat) NFGMH SEQ ID NO: 58 HC CDR2 (Kabat) YISSGSSTIYYADTLKG SEQ ID NO: 59 HC CDR3 (Kabat) RGEGAMDY SEQ ID NO: 60 HC CDR1 (Chothia) GFTFSNF SEQ ID NO: 61 HC CDR2 (Chothia) SSGSST SEQ ID NO: 62 HC CDR3 (Chothia) RGEGAMDY SEQ ID NO: 15 VH DVQLVESGGGLVQPGGSRKLSCAASGFTFSNFGMH WVRQAPDKGLEWVAYISSGSSTIYYADTLKGRFTIS RDNPKNTLFLQMTSLRSEDTAMYYCARRGEGAMD YWGQGTSVTVSS SEQ ID NO: 20 LC CDR1 (Combined) RASSSVNYIY SEQ ID NO: 21 LC CDR2 (Combined) YTSNLAP SEQ ID NO: 22 LC CDR3 (Combined) QQFTSSPFT SEQ ID NO: 63 LC CDR1 (Kabat) RASSSVNYIY SEQ ID NO: 64 LC CDR2 (Kabat) YTSNLAP SEQ ID NO: 65 LC CDR3 (Kabat) QQFTSSPFT SEQ ID NO: 66 LC CDR1 (Chothia) RASSSVNYIY SEQ ID NO: 67 LC CDR2 (Chothia) YTSNLAP SEQ ID NO: 68 LC CDR3 (Chothia) QQFTSSPFT SEQ ID NO: 16 VL ENVLTQSPAIMSASLGEKVTMSCRASSSVNYIYWY QQKSDASPKLWIYYTSNLAPGVPTRFSGSGSGNSYS LTISSMEGEDAATYYCQQFTSSPFTFGSGTKLEIK Antibody B humanized (B-H) Antibody B-H.1A HC-1 SEQ ID NO: 17 HC CDR1 (Combined) GFTFSNFGMH SEQ ID NO: 18 HC CDR2 (Combined) YISSGSSTIYYADTLKG SEQ ID NO: 19 HC CDR3 (Combined) RGEGAMDY SEQ ID NO: 3438 VH EVQLVESGGGLVQPGGSLRLSCAASGFTFSNFGMH WVRQAPGKGLEWVSYISSGSSTIYYADTLKGRFTIS RDNAKNSLYLQMNSLRAEDTAVYYCARRGEGAM DYWGQGTTVTVSS SEQ ID NO: 31 DNA VH GAGGTGCAGCTGGTTGAATCTGGCGGAGGATTG GTTCAGCCTGGCGGCTCTCTGAGACTGTCTTGTG CCGCTTCTGGCTTCACCTTCTCCAACTTCGGCATG CACTGGGTCCGACAGGCCCCTGGAAAAGGACTG GAATGGGTGTCCTACATCTCCTCCGGCTCCTCCA CCATCTACTACGCTGACACCCTGAAGGGCAGATT CACCATCTCTCGGGACAACGCCAAGAACTCCCTG TACCTGCAGATGAACAGCCTGAGAGCCGAGGAC ACCGCCGTGTACTACTGTGCTAGAAGAGGCGAG GGCGCCATGGATTATTGGGGCCAGGGAACCACA GTGACCGTGTCTAGC Antibody B-H.1B UC-2 SEQ ID NO: 17 HC CDR1 (Combined) GFTFSNFGMH SEQ ID NO: 18 HC CDR2 (Combined) YISSGSSTIYYADTLKG SEQ ID NO: 19 HC CDR3 (Combined) RGEGAMDY SEQ ID NO: 25 VH EVQLVESGGGLVQPGGSLRLSCAASGFTFSNFGMH WVRQAPGKGLEWVSYISSGSSTIYYADTLKGRFTIS RDNSKNTLYLQMNSLRAEDTAVYYCARRGEGAM DYWGQGTTVTVSS SEQ ID NO: 32 DNA VH GAGGTGCAGCTGGTTGAATCTGGCGGAGGATTG GTTCAGCCTGGCGGCTCTCTGAGACTGTCTTGTG CCGCTTCTGGCTTCACCTTCTCCAACTTCGGCATG CACTGGGTCCGACAGGCCCCTGGAAAAGGACTG GAATGGGTGTCCTACATCTCCTCCGGCTCCTCCA CCATCTACTACGCTGACACCCTGAAGGGCAGATT CACCATCAGCCGGGACAACTCCAAGAACACCCT GTACCTGCAGATGAACTCCCTGAGAGCCGAGGA CACCGCCGTGTACTACTGTGCTAGAAGAGGCGA GGGCGCCATGGATTATTGGGGCCAGGGAACCAC AGTGACCGTGTCTAGC Antibody B-H.1C HC-3 SEQ ID NO: 17 HC CDR1 (Combined) GFTFSNFGMH SEQ ID NO: 18 HC CDR2 (Combined) YISSGSSTIYYADTLKG SEQ ID NO: 19 HC CDR3 (Combined) RGEGAMDY SEQ ID NO: 23 VH QVQLVESGGGVVQPGRSLRLSCAASGFTFSNFGMH WVRQAPGKGLEWVAYISSGSSTIYYADTLKGRFTIS RDNSKNTLYLQMNSLRAEDTAVYYCARRGEGAM DYWGQGTTVTVSS SEQ ID NO: 33 DNA VH CAGGTGCAGCTGGTGGAATCTGGTGGCGGAGTT GTGCAGCCTGGCAGATCCCTGAGACTGTCTTGTG CCGCCTCTGGCTTCACCTTCTCCAACTTCGGCATG CACTGGGTCCGACAGGCCCCTGGAAAAGGATTG GAGTGGGTCGCCTACATCTCCTCCGGCTCCTCCA CCATCTACTACGCTGACACCCTGAAGGGCAGATT CACCATCAGCCGGGACAACTCCAAGAACACCCT GTACCTGCAGATGAACTCCCTGAGAGCCGAGGA CACCGCCGTGTACTACTGTGCTAGAAGAGGCGA GGGCGCCATGGATTATTGGGGCCAGGGAACCAC AGTGACCGTGTCTAGC Antibody B-H.1D LC-1 SEQ ID NO: 20 LC CDR1 (Combined) RASSSVNYIY SEQ ID NO: 21 LC CDR2 (Combined) YTSNLAP SEQ ID NO: 22 LC CDR3 (Combined) QQFTSSPFT SEQ ID NO: 26 VL DNQLTQSPSFLSASVGDRVTITCRASSSVNYIYWYQ QKPGKAPKLLIYYTSNLAPGVPSRFSGSGSGNEYTL TISSLQPEDFATYYCQQFTSSPFTFGQGTKLEIK SEQ ID NO: 34 DNA VL GATAACCAGCTGACCCAGTCTCCTAGCTTCCTGT CTGCCTCTGTGGGCGACAGAGTGACAATTACCTG CCGGGCCTCCTCCTCCGTGAACTACATCTACTGG TATCAGCAGAAGCCCGGCAAGGCCCCTAAGCTG TGCCCTCTAGATTTTCCGGATCTGGCTCCGGCAA CGAGTATACCCTGACAATCTCCAGCCTGCAGCCT GAGGACTTCGCCACCTACTACTGCCAGCAGTTCA CCTCCTCTCCATTCACCTTTGGCCAGGGCACCAA GCTGGAAATCAAA Antibody B-H.1E LC-2 SEQ ID NO: 20 LC CDR1 (Combined) RASSSVNYIY SEQ ID NO: 21 LC CDR2 (Combined) YTSNLAP SEQ ID NO: 22 LC CDR3 (Combined) QQFTSSPFT SEQ ID NO: 27 VL DNQLTQSPSSLSASVGDRVTITCRASSSVNYIYWYQ QKPGKAPKLLIYYTSNLAPGVPSRFSGSGSGNDYTL TISSLQPEDFATYYCQQFTSSPFTFGQGTKLEIK SEQ ID NO: 35 DNA VL ATAACCAGCTGACCCAGTCTCCTTCCAGCCTGTC TGCTTCTGTGGGCGACAGAGTGACAATTACCTGC CGGGCCTCCTCCTCCGTGAACTACATCTACTGGT ATCAGCAGAAGCCCGGCAAGGCCCCTAAGCTGC TGATCTACTACACCTCCAATCTGGCCCCTGGCGT GCCCTCTAGATTTTCCGGATCTGGCTCCGGCAAC GACTATACCCTGACAATCTCCAGCCTGCAGCCTG AGGACTTCGCCACCTACTACTGCCAGCAGTTCAC CTCCTCTCCATTCACCTTTGGCCAGGGCACCAAG CTGGAAATCAAA Antibody B-H.1F C-3 SEQ ID NO: 20 LC CDR1 (Combined) RASSSVNYIY SEQ ID NO: 21 LC CDR2 (Combined) YTSNLAP SEQ ID NO: 22 LC CDR3 (Combined) QQFTSSPFT SEQ ID NO: 28 VL ENVLTQSPATLSVSPGERATLSCRASSSVNYIYWYQ QKPGQAPRLLIYYTSNLAPGIPARFSGSGSGNEYTL TISSLQSEDFAVYYCQQFTSSPFTFGQGTKLEIK SEQ ID NO: 36 DNA VL GAGAATGTGCTGACCCAGTCTCCTGCCACACTGT CTGTTAGCCCTGGCGAGAGAGCTACCCTGAGCTG CAGAGCCTCTTCCTCCGTGAACTACATCTACTGG TATCAGCAGAAGCCCGGCCAGGCTCCTAGACTGC TGATCTACTACACCTCCAATCTGGCCCCTGGCAT CCCTGCCAGATTTTCCGGATCTGGCTCCGGCAAC GAGTATACCCTGACCATCTCCAGCCTGCAGTCCG AGGACTTTGCTGTGTACTATTGCCAGCAGTTCAC AAGCAGCCCTTTCACCTTTGGCCAGGGCACCAAG CTGGAAATCAAA Antibody B-H.1G LC-4 SEQ ID NO: 20 LC CDR1 (Combined) RASSSVNYIY SEQ ID NO: 21 LC CDR2 (Combined) YTSNLAP SEQ ID NO: 22 LC CDR3 (Combined) QQFTSSPFT SEQ ID NO: 29 VL QNVLTQPPSASGTPGQRVTISCRASSSVNYIYWYQQ LPGTAPKLLIYYTSNLAPGVPDRFSGSGSGNSYSLAI SGLRSEDEADYYCQQFTSSPFTFGTGTKVTVL SEQ ID NO: 37 DNA VL CAGAATGTGCTGACCCAACCTCCTTCCGCCTCTG GCACACCTGGACAGAGAGTGACAATCTCCTGCC GGGCCTCCTCCTCCGTGAACTACATCTACTGGTA TCAGCAGCTGCCCGGCACCGCTCCTAAACTGCTG ATCTACTACACCTCCAATCTGGCCCCTGGCGTGC CCGATAGATTTTCCGGATCTGGCTCCGGCAACTC CTACAGCCTGGCTATCTCTGGCCTGAGATCTGAG GACGAGGCCGACTACTACTGCCAGCAGTTCACCT CCTCTCCATTCACCTTTGGCACCGGCACCAAAGT GACAGTTCTT Antibody B-H.1H LC-5 SEQ ID NO: 20 LC CDR1 (Combined) RASSSVNYIY SEQ ID NO: 21 LC CDR2 (Combined) YTSNLAP SEQ ID NO: 22 LC CDR3 (Combined) QQFTSSPFT SEQ ID NO: 30 VL SNELTQPPSVSVSPGQTARITCRASSSVNYIYWYQQ KSGQAPVLVIYYTSNLAPGIPERFSGSGSGNMYTLT ISGAQVEDEADYYCQQFTSSPFTFGTGTKVTVL SEQ ID NO: 38 DNA VL TCTAATGAGCTGACCCAGCCTCCTTCCGTGTCCG TGTCTCCTGGACAGACCGCCAGAATTACCTGCCG GGCCTCCTCCTCCGTGAACTACATCTACTGGTAT CAGCAGAAGTCCGGCCAGGCTCCTGTGCTCGTGA TCTACTACACCTCCAATCTGGCCCCTGGCATCCC TGAGAGATTCTCCGGATCTGGCTCCGGCAACATG TACACCCTGACCATCTCTGGCGCCCAGGTGGAAG ATGAGGCCGACTACTACTGCCAGCAGTTCACCTC CTCTCCATTCACCTTTGGCACCGGCACCAAAGTG ACAGTTCTT Antibody B-H.1 SEQ ID NO: 3280 Chain 1: Fc only METDTLLLWVLLLWVPGSTGDKTHTCPPCPAPELL GGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDP EVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSV LTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK GQPREPQVYTLPPCREEMTKNQVSLWCLVKGFYPS DIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT VDKSRWQQGNVFSCSVMHEALHNRFTQKSLSLSP GK SEQ ID NO: 3281 Chain2: humanized B-H METDTLLLWVLLLWVPGSTGEVQLVESGGGLVQP scFv GGSLRLSCAASGFTFSNFGMHWVRQAPGKGLEWV SYISSGSSTIYYADTLKGRFTISRDNSKNTLYLQMNS LRAEDTAVYYCARRGEGAMDYWGQGTTVTVSSG GGGSGGGGSGGGGSGGGGSDNQLTQSPSFLSASVG DRVTITCRASSSVNYIYWYQQKPGKAPKLLIYYTSN LAPGVPSRFSGSGSGNEYTLTISSLQPEDFATYYCQ QFTSSPFTFGQGTKLEIKGGGGSDKTHTCPPCPAPE LLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE DPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVV SVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISK AKGQPREPQVCTLPPSREEMTKNQVSLSCAVKGFY PSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSK LTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSL SPGKGGGGSGGGGSGLNDIFEAQKIEWHE SEQ ID NO: 3642 scFv EVQLVESGGGLVQPGGSLRLSCAASGFTFSNFGMH WVRQAPGKGLEWVSYISSGSSTIYYADTLKGRFTIS RDNSKNTLYLQMNSLRAEDTAVYYCARRGEGAM DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DNQLTQSPSFLSASVGDRVTITCRASSSVNYIYWYQ QKPGKAPKLLIYYTSNLAPGVPSRFSGSGSGNEYTL TISSLQPEDFATYYCQQFTSSPFTFGQGTKLEIK Antibody B-H.2 SEQ ID NO: 1338 scFv EVQLVESGGGLVQPGGSLRLSCAASGFTFSNFGMH WVRQAPGKGLEWVSYISSGSSTIYYADTLKGRFTIS RDNSKNTLYLQMNSLRAEDTAVYYCARRGEGAM DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DNQLTQSPSSLSASVGDRVTITCRASSSVNYIYWYQ QKPGKAPKLLIYYTSNLAPGVPSRFSGSGSGNDYTL TISSLQPEDFATYYCQQFTSSPFTFGQGTKLEIK Antibody B-H.3 SEQ ID NO: 1339 scFv EVQLVESGGGLVQPGGSLRLSCAASGFTFSNFGMH WVRQAPGKGLEWVSYISSGSSTIYYADTLKGRFTIS RDNSKNTLYLQMNSLRAEDTAVYYCARRGEGAM DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSS NELTQPPSVSVSPGQTARITCRASSSVNYIYWYQQK SGQAPVLVIYYTSNLAPGIPERFSGSGSGNMYTLTIS GAQVEDEADYYCQQFTSSPFTFGTGTKVTVL Antibody B-H.4 SEQ ID NO: 1340 scFv QVQLVESGGGVVQPGRSLRLSCAASGFTFSNFGMH WVRQAPGKGLEWVAYISSGSSTIYYADTLKGRFTIS RDNSKNTLYLQMNSLRAEDTAVYYCARRGEGAM DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DNQLTQSPSFLSASVGDRVTITCRASSSVNYIYWYQ QKPGKAPKLLIYYTSNLAPGVPSRFSGSGSGNEYTL TISSLQPEDFATYYCQQFTSSPFTFGQGTKLEIK Antibody B-H.5 SEQ ID NO: 1341 scFv QVQLVESGGGVVQPGRSLRLSCAASGFTFSNFGMH WVRQAPGKGLEWVAYISSGSSTIYYADTLKGRFTIS RDNSKNTLYLQMNSLRAEDTAVYYCARRGEGAM DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGS DNQLTQSPSSLSASVGDRVTITCRASSSVNYIYWYQ QKPGKAPKLLIYYTSNLAPGVPSRFSGSGSGNDYTL TISSLQPEDFATYYCQQFTSSPFTFGQGTKLEIK Antibody B-H.6 SEQ ID NO: 1342 scFv QVQLVESGGGVVQPGRSLRLSCAASGFTFSNFGMH WVRQAPGKGLEWVAYISSGSSTIYYADTLKGRFTIS RDNSKNTLYLQMNSLRAEDTAVYYCARRGEGAM DYWGQGTTVTVSSGGGGSGGGGSGGGGSGGGGSS NELTQPPSVSVSPGQTARITCRASSSVNYIYWYQQK SGQAPVLVIYYTSNLAPGIPERFSGSGSGNMYTLTIS GAQVEDEADYYCQQFTSSPFTFGTGTKVTVL

TABLE 3 Constant region amino acid sequences of human IgG heavy chains and human kappa light chain Human kappa LC RTVAAPSVFI FPPSDEQLKS GTASVVCLLN NFYPREAKVQ constant region WKVDNALQSG NSQESVTEQD SKDSTYSLSS TLTLSKADYE SEQ ID NO: 39 KHKVYACEVT HQGLSSPVTK SFNRGEC IgG4 (S228P) HC ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSG mutant constant VHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKR region (EU VESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVD Numbering) VSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDW SEQ ID NO: 40 LNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQ VSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLT VDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLG IgG1 wild type HC ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG SEQ ID NO: 41 VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKRV EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVD VSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQ DWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMT KNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLY SKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK IgG1 (N297A) HC ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG mutant constant VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKRV region (EU EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVD Numbering) VSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQ SEQ ID NO: 42 DWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMT KNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLY SKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK IgM constant HC GSASAPTLFPLVSCENSPSDTSSVAVGCLAQDFLPDSITFSWKYKNNS delta CDC DISSTRGFPSVLRGGKYAATSQVLLPSKDVMQGTDEHVVCKVQHPNGNK (P311A, P313S) EKNVPLPVIAELPPKVSVFVPPRDGFFGNPRKSKLICQATGFSPRQIQ SEQ ID NO: 73 VSWLREGKQVGSGVTTDQVQAEAKESGPTTYKVTSTLTIKESDWLGQSM FTCRVDHRGLTFQQNASSMCVPDQDTAIRVFAIPPSFASIFLTKSTKLT CLVTDLTTYDSVTISWTRQNGEAVKTHTNISESHPNATFSAVGEASICE DDWNSGERFTCTVTHTDLASSLKQTISRPKGVALHRPDVYLLPPAREQL NLRESATITCLVTGFSPADVFVQWMQRGQPLSPEKYVTSAPMPEPQAP GRYFAHSILTVSEEEWNTGETYTCVVAHEALPNRVTERTVDKSTGKPT LYNVSLVMSDTAGTCY IgGA1 HC ASPTSPKVFPLSLCSTQPDGNVVIACLVQGFFPQEPLSVTWSESGQGVT SEQ ID NO: 74 ARNFPPSQDASGDLYTTSSQLTLPATQCLAGKSVTCHVKHYTNPSQDVT VPCPVPSTPPTPSPSTPPTPSPSCCHPRLSLHRPALEDLLLGSEANLTC TLTGLRDASGVTFTWTPSSGKSAVQGPPERDLCGCYSVSSVLPGCAEPW NHGKTFTCTAAYPESKTPLTATLSKSGNTFRPEVHLLPPPSEELALNEL VTLTCLARGFSPKDVLVRWLQGSQELPREKYLTWASRQEPSQGTTTFA VTSILRVAAEDWKKGDTFSCMVGHEALPLAFTQKTIDRLAGKPTHVN VSVVMAEVDGTCY IgGA2 HC ASPTSPKVFPLSLDSTPQDGNVVVACLVQGFFPQEPLSVTWSESGQNV SEQ ID NO: 75 TARNFPPSQDASGDLYTTSSQLTLPATQCPDGKSVTCHVKHYTNSSQD VTVPCRVPPPPPCCHPRLSLHRPALEDLLLGSEANLTCTLTGLRDASG ATFTWTPSSGKSAVQGPPERDLCGCYSVSSVLPGCAQPWNHGETFTCT AAHPELKTPLTANITKSGNTFRPEVHLLPPPSEELALNELVTLTCLAR GFSPKDVLVRWLQGSQELPREKYLTWASRQEPSQGTTTYAVTSILRVA AEDWKKGETFSCMVGHEALPLAFTQKTIDRMAGKPTHINVSVVMAEAD GTCY Human Ig_J HC MKNHLLFWGVLAVFIKAVHVKAQEDERIVLVDNKCKCARITSRIIRSSE chain DPNEDIVERNIRIIVPLNNRENISDPTSPLRTRFVYHLSDLCKKCDPTE SEQ ID NO: 76 VELDNQIVTATQSNICDEDSATETCYTYDRNKCYTAVVPLVYGGETKMV ETALTPDACYPD

Anti-TCRβ V5 Antibodies

Accordingly, in one aspect, the disclosure provides an anti-TCRβV antibody molecule that binds to human TCRβ V5. In some embodiments, the TCRβ V5 subfamily comprises TCRβ V5-5*01, TCRβ V5-6*01, TCRβ V5-4*01, TCRβ V5-8*01, TCRβ V5-1*01, or a variant thereof.

Exemplary anti-TCRβ V5 antibodies of the disclosure are provided in Table 10. In some embodiments, the anti-TCRβ V5 is antibody C, e.g., humanized antibody C (antibody C-H), as provided in Table 10. In some embodiments, the anti-TCRβV antibody comprises one or more (e.g., all three) of a LC CDR1, LC CDR2, and LC CDR3 provided in Table 10; and/or one or more (e.g., all three) of a HC CDR1, HC CDR2, and HC CDR3 provided in Table 10, or a sequence with at least 95% identity thereto. In some embodiments, antibody C comprises a variable heavy chain (VH) and/or a variable light chain (VL) provided in Table 10, or a sequence with at least 95% identity thereto.

TABLE 10 Amino acid sequences for anti TCRβ V5 antibodies Amino acid and nucleotide sequences for murine and humanized antibody molecules which bind to TCRVB 5 (e.g., TCRVB 5-5 or TCRVB 5-6). The amino acid the heavy and light chain CDRs, and the amino acid and nucleotide sequences of the heavy and light chain variable regions, and the heavy and light chains are shown. Murine antibody C, also referred to as 4H11 SEQ ID NO: 1315 HC CDR1 (Kabat) AYGVN SEQ ID NO: 1316 HC CDR2 (Kabat) MIWGDGNTDYNSALKS SEQ ID NO: 1317 HC CDR3 (Kabat) DRVTATLYAMDY SEQ ID NO: 1318 HC CDR1 (Chothia) GFSLTAY SEQ ID NO: 1319 HC CDR2 (Chothia) WGDGN SEQ ID NO: 1317 HC CDR3 (Chothia) DRVTATLYAMDY SEQ ID NO: 1320 HC CDR1 (Combined) GFSLTAYGVN SEQ ID NO: 1316 HC CDR2 (Combined) MIWGDGNTDYNSALKS SEQ ID NO: 1317 HC CDR3 (Combined) DRVTATLYAMDY SEQ ID NO: 1321 LC CDR1 (Kabat) SASQGISNYLN SEQ ID NO: 1322 LC CDR2 (Kabat) YTSSLHS SEQ ID NO: 1323 LC CDR3 (Kabat) QQYSKLPRT SEQ ID NO: 1321 LC CDR1 (Chothia) SASQGISNYLN SEQ ID NO: 1322 LC CDR2 (Chothia) YTSSLHS SEQ ID NO: 1323 LC CDR3 (Chothia) QQYSKLPRT SEQ ID NO: 1321 LC CDR1 (Combined) SASQGISNYLN SEQ ID NO: 1322 LC CDR2 (Combined) YTSSLHS SEQ ID NO: 1323 LC CDR3 (Combined) QQYSKLPRT SEQ ID NO: 232 VH DIQMTQTTSSLSASLGDRVTISCSASQGISNYLNW YQQKPDGTVKLLIYYTSSLHSGVPSRFSGSGSGTD YSLTISNLEPEDIATYYCQQYSKLPRTFGGGTKVE IK SEQ ID NO: 233 VL QVQLKESGPGLVAPSQSLSITCTVSGFSLTAYGVN WVRQPPGKGLEWLGMIWGDGNTDYNSALKSRL SISKDNSKSQVFLKMNSLQTDDTARYYCARDRV TATLYAMDYWGQGTSVTVSS Humanized antibody C C-H-1 antibody SEQ ID NO: 1315 HC CDR1 (Kabat) AYGVN SEQ ID NO: 1316 HC CDR2 (Kabat) MIWGDGNTDYNSALKS SEQ ID NO: 1317 HC CDR3 (Kabat) DRVTATLYAMDY SEQ ID NO: 1318 HC CDR1 (Chothia) GFSLTAY SEQ ID NO: 1319 HC CDR2 (Chothia) WGDGN SEQ ID NO: 1317 HC CDR3 (Chothia) DRVTATLYAMDY SEQ ID NO: 1320 HC CDR1 (Combined) GFSLTAYGVN SEQ ID NO: 1316 HC CDR2 (Combined) MIWGDGNTDYNSALKS SEQ ID NO: 1317 HC CDR3 (Combined) DRVTATLYAMDY SEQ ID NO: 1321 LC CDR1 (Kabat) SASQGISNYLN SEQ ID NO: 1322 LC CDR2 (Kabat) YTSSLHS SEQ ID NO: 1323 LC CDR3 (Kabat) QQYSKLPRT SEQ ID NO: 1321 LC CDR1 (Chothia) SASQGISNYLN SEQ ID NO: 1322 LC CDR2 (Chothia) YTSSLHS SEQ ID NO: 1323 LC CDR3 (Chothia) QQYSKLPRT SEQ ID NO: 1321 LC CDR1 (Combined) SASQGISNYLN SEQ ID NO: 1322 LC CDR2 (Combined) YTSSLHS SEQ ID NO: 1323 LC CDR3 (Combined) QQYSKLPRT SEQ ID NO: 1324 VL DIQMTQSPSSLSASVGDRVTITCSASQGISNYLNW YQQTPGKAPKLLIYYTSSLHSGVPSRFSGSGSGTD YTFTISSLQPEDIATYYCQQYSKLPRTFGQGTKLQ IT SEQ ID NO: 1325 VH QVQLQESGPGLVRPSQTLSLTCTVSGFSLTAYGV NWVRQPPGRGLEWLGMIWGDGNTDYNSALKSR VTMLKDTSKNQFSLRLSSVTAADTAVYYCARDR VTATLYAMDYWGQGSLVTVSS Humanized antibody C Variable light chain (VL) SEQ ID VL C-H- DIQMTQSPSFLSASVGDRVTITCSASQGISNYLNWYQQKPGKAVKL NO: 3000 VL.1 LIYYTSSLHSGVPSRFSGSGSGTEYTLTISSLQPEDFATYYCQQYS KLPRTFGGGTKVEIK SEQ ID VL C-H- DIQMTQSPSSLSASVGDRVTITCSASQGISNYLNWYQQKPGKAVKL NO: 3001 VL.2 LIYYTSSLHSGVPSRFSGSGSGTDYTLTISSLQPEDFATYYCQQYS KLPRTFGGGTKVEIK SEQ ID VL C-H- DIQMTQSPSSLSASVGDRVTITCSASQGISNYLNWYQQKPGKVVKL NO: 3002 VL.3 LIYYTSSLHSGVPSRFSGSGSGTDYTLTISSLQPEDVATYYCQQYS KLPRTFGGGTKVEIK SEQ ID VL C-H- DIQMTQSPSSLSASVGDRVTITCSASQGISNYLNWYQQKPGQAVKL NO: 3003 VL.4 LIYYTSSLHSGVPSRFSGSGSGTDYTLTISSLQPEDVATYYCQQYS KLPRTFGGGTKVEIK SEQ ID VL C-H- DIQMTQSPSSLSASVGDRVTITCSASQGISNYLNWYQQKPGKAVKL NO: 3004 VL.5 LIYYTSSLHSGVPSRFSGSGSGTDYTFTISSLQPEDIATYYCQQYSK LPRTFGGGTKVEIK SEQ ID VL C-H- DIQMTQSPSSLSASVGDRVTITCSASQGISNYLNWYQQKPGKTVKLL NO: 3005 VL.6 IYYTSSLHSGIPSRFSGSGSGTDYTLTIRSLQPEDFATYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- AIQMTQSPSSLSASVGDRVTITCSASQGISNYLNWYQQKPGKAVKL NO: 3006 VL.7 LIYYTSSLHSGVPSRFSGSGSGTDYTLTISSLQPEDFATYYCQQYSKL PRTFGGGTKVEIK SEQ ID VL C-H- DIQMTQSPSSVSASVGDRVTITCSASQGISNYLNWYQQKPGKAVKL NO: 3007 VL.8 LIYYTSSLHSGVPSRFSGSGSGTDYTLTISSLQPEDFATYYCQQYSKL PRTFGGGTKVEIK SEQ ID VL C-H- DIQMTQSPSSLSASVGDRVTITCSASQGISNYLNWYQQKPGKAVKR NO: 3008 VL.9 LIYYTSSLHSGVPSRFSGSGSGTEYTLTISNLQPEDFATYYCQQYSKL PRTFGGGTKVEIK SEQ ID VL C-H- AIRMTQSPFSLSASVGDRVTITCSASQGISNYLNWYQQKPAKAVKLF NO: 3009 VL.10 IYYTSSLHSGVPSRFSGSGSGTDYTLTISSLQPEDFATYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- DIQMTQSPSSLSASVGDRVTITCSASQGISNYLNWYQQKPGKAVKR NO: 3010 VL.11 LIYYTSSLHSGVPSRFSGSGSGTEYTLTISSLQPEDFATYYCQQYSKL PRTFGGGTKVEIK SEQ ID VL C-H- DIQMTQSPSTLSASVGDRVTITCSASQGISNYLNWYQQKPGKAVKL NO: 3011 VL.12 LIYYTSSLHSGVPSRFSGSGSGTEYTLTISSLQPDDFATYYCQQYSKL PRTFGGGTKVEIK SEQ ID VL C-H- DIQMTQSPSSLSASVGDRVTITCSASQGISNYLNWYQQKPGKAVKSL NO:3012 VL.13 IYYTSSLHSGVPSRFSGSGSGTDYTLTISSLQPEDFATYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- DIQMTQSPSSLSASVGDRVTITCSASQGISNYLNWYQQKPGKAVKSL NO: 3013 VL.14 IYYTSSLHSGVPSKFSGSGSGTDYTLTISSLQPEDFATYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- DIQMTQSPSSLSASVGDRVTITCSASQGISNYLNWYQQKPEKAVKSL NO: 3014 VL.15 IYYTSSLHSGVPSRFSGSGSGTDYTLTISSLQPEDFATYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- DIQMTQSPSAMSASVGDRVTITCSASQGISNYLNWYQQKPGKVVKR NO: 3015 VL.16 LIYYTSSLHSGVPSRFSGSGSGTEYTLTISSLQPEDFATYYCQQYSKL PRTFGGGTKVEIK SEQ ID VL C-H- DIVMTQSPDSLAVSLGERATINCSASQGISNYLNWYQQKPGQPVKL NO: 3016 VL.17 LIYYTSSLHSGVPDRFSGSGSGTDYTLTISSLQAEDVAVYYCQQYSK LPRTFGGGTKVEIK SEQ ID VL C-H- EIVMTQSPGTLSLSPGERATLSCSASQGISNYLNWYQQKPGQAVKLL NO: 3017 VL.18 IYYTSSLHSGIPDRFSGSGSGTDYTLTISRLEPEDFAVYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- EIVMTQSPPTLSLSPGERVTLSCSASQGISNYLNWYQQKPGQAVKLL NO: 3018 VL.19 IYYTSSLHSGIPARFSGSGSGTDYTLTISSLQPEDFAVYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- EIVMTQSPPTLSLSPGERVTLSCSASQGISNYLNWYQQKPGQAVKLL NO: 3019 VL.20 IYYTSSLHSSIPARFSGSGSGTDYTLTISSLQPEDFAVYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- EIVMTQSPATLSLSPGERATLSCSASQGISNYLNWYQQKPGQAVKLL NO: 3020 VL.21 IYYTSSLHSGIPARFSGSGSGTDYTLTISSLEPEDFAVYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- EIVMTQSPATLSLSPGERATLSCSASQGISNYLNWYQQKPGQAVKLL NO: 3021 VL.22 IYYTSSLHSGIPARFSGSGSGTDYTLTISRLEPEDFAVYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- EIVMTQSPATLSLSPGERATLSCSASQGISNYLNWYQQKPGQAVKLL NO: 3022 VL.23 IYYTSSLHSGIPDRFSGSGSGTDYTLTISRLEPEDFAVYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- EIVMTQSPATLSLSPGERATLSCSASQGISNYLNWYQQKPGLAVKLL NO: 3023 VL.24 IYYTSSLHSGIPDRFSGSGSGTDYTLTISRLEPEDFAVYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- DIQMIQSPSFLSASVGDRVSIICSASQGISNYLNWYLQKPGKSVKLFI NO: 3024 VL.25 YYTSSLHSGVSSRFSGRGSGTDYTLTIISLKPEDFAAYYCQQYSKLPR TFGGGTKVEIK SEQ ID VL C-H- EIVMTQSPATLSLSPGERATLSCSASQGISNYLNWYQQKPGQAVKLL NO: 3025 VL.26 IYYTSSLHSGIPARFSGSGSGTDYTLTISSLQPEDFAVYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- EIVMTQSPATLSLSPGERATLSCSASQGISNYLNWYQQKPGQAVKLL NO: 3026 VL.27 IYYTSSLHSGIPARFSGSGPGTDYTLTISSLEPEDFAVYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- DIVMTQTPLSLSVTPGQPASISCSASQGISNYLNWYLQKPGQSVKLLI NO: 3027 VL.28 YYTSSLHSGVPDRFSGSGSGTDYTLKISRVEAEDVGVYYCQQYSKL PRTFGGGTKVEIK SEQ ID VL C-H- DIVMTQTPLSLSVTPGQPASISCSASQGISNYLNWYLQKPGQPVKLLI NO: 3028 VL.29 YYTSSLHSGVPDRFSGSGSGTDYTLKISRVEAEDVGVYYCQQYSKL PRTFGGGTKVEIK SEQ ID VL C-H- DIVMTQSPAFLSVTPGEKVTITCSASQGISNYLNWYQQKPDQAVKL NO: 3029 VL.30 LIYYTSSLHSGVPSRFSGSGSGTDYTFTISSLEAEDAATYYCQQYSKL PRTFGGGTKVEIK SEQ ID VL C-H- DIVMTQSPLSLPVTPGEPASISCSASQGISNYLNWYLQKPGQSVKLLI NO: 3030 VL.31 YYTSSLHSGVPDRFSGSGSGTDYTLKISRVEAEDVGVYYCQQYSKL PRTFGGGTKVEIK SEQ ID VL C-H- DIVMTQTPLSLPVTPGEPASISCSASQGISNYLNWYLQKPGQSVKLLI NO: 3031 VL.32 YYTSSLHSGVPDRFSGSGSGTDYTLKISRVEAEDVGVYYCQQYSKL PRTFGGGTKVEIK SEQ ID VL C-H- EIVMTQSPATLSVSPGERATLSCSASQGISNYLNWYQQKPGQAVKL NO: 3032 VL.33 LIYYTSSLHSGIPARFSGSGSGTEYTLTISILQSEDFAVYYCQQYSK LPRTFGGGTKVEIK SEQ ID VL C-H- EIVMTQSPATLSVSPGERATLSCSASQGISNYLNWYQQKPGQAVKL NO: 3033 VL.34 LIYYTSSLHSGIPARFSGSGSGTEYTLTISSLQSEDFAVYYCQQYSKL PRTFGGGTKVEIK SEQ ID VL C-H- DIVMTQSPLSLPVTLGQPASISCSASQGISNYLNWYQQRPGQSVKRLI NO: 3034 VL.35 YYTSSLHSGVPDRFSGSGSGTDYTLKISRVEAEDVGVYYCQQYSKL PRTFGGGTKVEIK SEQ ID VL C-H- EITMTQSPAFMSATPGDKVNISCSASQGISNYLNWYQQKPGEAVKFI NO: 3035 VL.36 IYYTSSLHSGIPPRFSGSGYGTDYTLTINNIESEDAAYYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- DIVMTQTPLSSPVTLGQPASISCSASQGISNYLNWYQQRPGQPVKLLI NO: 3036 VL.37 YYTSSLHSGVPDRFSGSGAGTDYTLKISRVEAEDVGVYYCQQYSKL PRTFGGGTKVEIK SEQ ID VL C-H- EIVMTQSPDFQSVTPKEKVTITCSASQGISNYLNWYQQKPDQSVKLL NO: 3037 VL.38 IYYTSSLHSGVPSRFSGSGSGTDYTLTINSLEAEDAATYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- EIVMTQTPLSLSITPGEQASISCSASQGISNYLNWYLQKARPVVKLLI NO: 3038 VL.39 YYTSSLHSGVPDRFSGSGSGTDYTLKISRVEAEDFGVYYCQQYSKLP RTFGGGTKVEIK SEQ ID VL C-H- EIVMTQTPLSLSITPGEQASMSCSASQGISNYLNWYLQKARPVVKLL NO: 3039 VL.40 IYYTSSLHSGVPDRFSGSGSGTDYTLKISRVEAEDFGVYYCQQYSKL PRTFGGGTKVEIK Humanized antibody C Variable HEAVY chain (VH) SEQ ID VH C-H- QVTLKESGPVLVKPTETLTLTCTVSGFSLTAYGVNWVRQPPGKALE NO: 3040 VH.1 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVVLTMTNMDPVDT ATYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVTLKESGPALVKPTETLTLTCTVSGFSLTAYGVNWVRQPPGKALE NO: 3041 VH.2 WLGMIWGDGNTDYNSALKSRLIISKDNSKSQVVLTMTNMDPVDTA TYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVTLKESGPALVKPTQTLTLTCTVSGFSLTAYGVNWVRQPPGKALE NO: 3042 VH.3 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVVLTMTNMDPVDT ATYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSGTLSLTCAVSGFSLTAYGVNWVRQPPGKGLE NO: 3043 VH.4 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVTLKESGPTLVKPTQTLTLTCTVSGFSLTAYGVNWVRQPPGKALE NO: 3044 VH.5 WLGMIWGDGNTDYNSALKSRLTITKDNSKSQVVLTMTNMDPVDT ATYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVTLKESGPALVKPTQTLTLTCTVSGFSLTAYGVNWVRQPPGKALE NO: 3045 VH.6 WLGMIWGDGNTDYNSALKSRLTITKDNSKSQVVLTMTNMDPVDT ATYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSQTLSLTCTVSGFSLTAYGVNWVRQPPGKGLE NO: 3046 VH.7 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSETLSLTCTVSGFSLTAYGVNWVRQPPGKGLE NO: 3047 VH.8 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSQTLSLTCAVSGFSLTAYGVNWVRQPPGKGLE NO: 3048 VH.9 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSDTLSLTCTVSGFSLTAYGVNWVRQPPGKGLE NO: 3049 VH.10 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSQTLSLTCTVSGFSLTAYGVNWVRQHPGKGLE NO: 3050 VH.11 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSQTLSLTCTVSGFSLTAYGVNWVRQPAGKGLE NO: 3051 VH.12 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSQTLSLTCAVSGFSLTAYGVNWVRQPPGKGLE NO: 3052 VH.13 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTAVDTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSETLSLTCTVSGFSLTAYGVNWVRQPPGKGLE NO: 3053 VH.14 WLGMIWGDGNTDYNSALKSRLTISKDNSKSHVSLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSETLSLTCAVSGFSLTAYGVNWVRQPPGKGLE NO: 3054 VH.15 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSQTLSLTCAVYGFSLTAYGVNWVRQPPGKGL NO: 3055 VH.16 EWLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- RVQLQESGPGLVKPSETLSLTCTVSGFSLTAYGVNWVRQPPGKGLE NO: 3056 VH.17 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVPLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSQTLSLTCTVSGFSLTAYGVNWVRQHPGKGLE NO: 3057 VH.18 WLGMIWGDGNTDYNSALKSLLTISKDNSKSQVSLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSDTLSLTCAVSGFSLTAYGVNWVRQPPGKGLE NO: 3058 VH.19 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTALDTAV YYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSDTLSLTCAVSGFSLTAYGVNWVRQPPGKGLE NO: 3059 VH.20 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTAVDTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGSGLVKPSQTLSLTCAVSGFSLTAYGVNWVRQPPGKGLE NO: 3060 VH.21 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- EVQLVESGGGLVQPGRSLRLSCTVSGFSLTAYGVNWVRQAPGKGL NO: 3061 VH.22 EWLGMIWGDGNTDYNSALKSRLTISKDNSKSIVYLQMNSLKTEDTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- EVQLVESGGGLVQPGPSLRLSCTVSGFSLTAYGVNWVRQAPGKGLE NO: 3062 VH.23 WLGMIWGDGNTDYNSALKSRLTISKDNSKSIVYLQMNSLKTEDTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGSGLVKPSQTLSLTCAVSGFSLTAYGVNWVRQSPGKGLE NO: 3063 VH.24 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSETLSLTCTVSGFSLTAYGVNWVRQPAGKGLE NO: 3064 VH.25 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- EVQLVESGGGLVKPGRSLRLSCTVSGFSLTAYGVNWVRQAPGKGL NO: 3065 VH.26 EWLGMIWGDGNTDYNSALKSRLTISKDNSKSIVYLQMNSLKTEDTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSETLSLTCAVYGFSLTAYGVNWVRQPPGKGLE NO: 3066 VH.27 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVYLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQESGPGLVKPSDTLSLTCAVSGFSLTAYGVNWVRQPPGKGLE NO: 3067 VH.28 WLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTAVDTG VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- EVQLVESGGGLVQPGGSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3068 VH.29 EWLGMIWGDGNTDYNSALKSRLTISKDNSKSSVYLQMNSLKTEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- EVQLVESGGGLVKPGGSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3069 VH.30 EWLGMIWGDGNTDYNSALKSRLTISKDNSKSTVYLQMNSLKTEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQQSGPGLVKPSQTLSLTCAVSGFSLTAYGVNWVRQSPSRGLE NO: 3070 VH.31 WLGMIWGDGNTDYNSALKSRLTINKDNSKSQVSLQLNSVTPEDTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLVESGGGLVQPGGSLRLSCSVSGFSLTAYGVNWVRQAPGKGL NO: 3071 VH.32 EWLGMIWGDGNTDYNSALKSRLTISKDNSKSTVYLQMNSLRAEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLQQWGAGLLKPSETLSLTCAVYGFSLTAYGVNWVRQPPGKGL NO: 3072 VH.33 EWLGMIWGDGNTDYNSALKSRLTISKDNSKSQVSLKLSSVTAADTA VYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLVESGGGVVQPGRSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3073 VH.34 EWLGMIWGDGNTDYNSALKSRLTISKDNSTSTVFLQMNSLRAEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- EVQLVESGGGLVQPGGSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3074 VH.35 EWLGMIWGDGNTDYNSALKSRLTISKDNSKSTVYLQMNSLRAEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- EVQLVESGGGLVQPGGSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3075 VH.36 EWLGMIWGDGNTDYNSALKSRLTISKDNAKSSVYLQMNSLRDEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- EVQLLESGGGLVQPGGSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3076 VH.37 EWLGMIWGDGNTDYNSALKSRLTISKDNSKSTVYLQMNSLRAEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLVESGGGLVKPGGSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3077 VH.38 EWLGMIWGDGNTDYNSALKSRLTISKDNAKSSVYLQMNSLRAEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- EVQLVESGGGLVQPGGSLKLSCAVSGFSLTAYGVNWVRQASGKGL NO: 3078 VH.39 EWLGMIWGDGNTDYNSALKSRLTISKDNSKSTVYLQMNSLKTEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLLESGGGLVKPGGSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3079 VH.40 EWLGMIWGDGNTDYNSALKSRLTISKDNAKSSVYLQMNSLRAEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLVESGGGVVQPGRSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3080 VH.41 EWLGMIWGDGNTDYNSALKSRLTISKDNSKSTVYLQMNSLRAEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLVESGGGVVQPGRSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3081 VH.42 EWLGMIWGDGNTDYNSALKSRLTISKDNSKSRVYLQMNSLRAEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLVESGGGVVQPGRSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3082 VH.43 EWLGMIWGDGNTDYNSALKSRLAISKDNSKSTVYLQMNSLRAEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- QVQLVESGGGVVQPGGSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3083 VH.44 EWLGMIWGDGNTDYNSALKSRLTISKDNSKSTVYLQMNSLRAEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- EVQLVESGGGLVQPGGSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3084 VH.45 EWLGMIWGDGNTDYNSALKSRLTISKDNAKSTVYLQMNSLRAEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- EVQLVESGGGLVQPGGSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3085 VH.46 EWLGMIWGDGNTDYNSALKSRLTISKDNAKSSVYLQMNSLRAEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- EVQLVESGGVVVQPGGSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3086 VH.47 EWLGMIWGDGNTDYNSALKSRLTISKDNSKSSVYLQMNSLRTEDT ALYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- EVQLVESGGGLVQPGGSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3087 VH.48 EWLGMIWGDGNTDYNSALKSRLTISKHNSKSTVYLQMNSLRAEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- EVQLVESGGGLVKPGGSLRLSCAVSGFSLTAYGVNWVRQAPGKGL NO: 3088 VH.49 EWLGMIWGDGNTDYNSALKSRLTISKDNAKSSVYLQMNSLRAEDT AVYYCARDRVTATLYAMDYWGQGTLVTVSS SEQ ID VH C-H- EVQLVESGGGLIQPGGSLRLSCAVSGFSLTAYGVNWVRQPPGKGLE NO: 3089 VH.50 WLGMIWGDGNTDYNSALKSRLTISKDNSKSTVYLQMNSLRAEDTA VYYCARDRVTATLYAMDYWGQGTLVTVSS

Exemplary anti-TCRβ V5 antibodies of the disclosure are provided in Table 11. In some embodiments, the anti-TCRβ V5 is antibody E, e.g., humanized antibody E (antibody E-H), as provided in Table 11. In some embodiments, the anti-TCRβV antibody comprises one or more (e.g., all three) of a LC CDR1, LC CDR2, and LC CDR3 provided in Table 11; and/or one or more (e.g., all three) of a HC CDR1, HC CDR2, and HC CDR3 provided in Table 11, or a sequence with at least 95% identity thereto. In some embodiments, antibody E comprises a variable heavy chain (VH) and/or a variable light chain (VL) provided in Table 11, or a sequence with at least 95% identity thereto.

In some embodiments, antibody E comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 3284 and/or a light chain comprising the amino acid sequence of SEQ ID NO: 3285, or a sequence with at least 95% identity thereto.

TABLE 11 Amino acid sequences for anti TCRβ V5 antibodies Amino acid and nucleotide sequences for murine and humanized antibody molecules which bind to TCRVB 5 (e.g., TCRVB 5-5 or TCRVB 5-6). The amino acid the heavy and light chain CDRs,  and the amino acid and nucleotide sequences of the heavy and light chain variable regions,  and the heavy and light chains are shown. Murine antibody E, also referred to as MH3-2 SEQ ID NO: 1298 HC CDR1 (Kabat) SSWMN SEQ ID NO: 1299 HC CDR2 (Kabat) RIYPGDGDTKYNGKFKG SEQ ID NO: 1300 HC CDR3 (Kabat) RGTGGWYFDV SEQ ID NO: 1302 HC CDR1 (Chothia) GYAFSSS SEQ ID NO: 1303 HC CDR2 (Chothia) YPGDGD SEQ ID NO: 1301 HC CDR3 (Chothia) RGTGGWYFDV SEQ ID NO: 1304 HC CDR1 (Combined) GYAFSSSWMN SEQ ID NO: 1299 HC CDR2 (Combined) RIYPGDGDTKYNGKFKG SEQ ID NO: 1301 HC CDR3 (Combined) RGTGGWYFDV SEQ ID NO: 1305 LC CDR1 (Kabat) RASESVDSSGNSFMH SEQ ID NO: 1306 LC CDR2 (Kabat) RASNLES SEQ ID NO: 1307 LC CDR3 (Kabat) QQSFDDPFT SEQ ID NO: 1308 LC CDR1 (Chothia) SESVDSSGNSF SEQ ID NO: 1306 LC CDR2 (Chothia) RASNLES SEQ ID NO: 1307 LC CDR3 (Chothia) QQSFDDPFT SEQ ID NO: 1305 LC CDR1 (Combined) RASESVDSSGNSFMH SEQ ID NO: 1306 LC CDR2 (Combined) RASNLES SEQ ID NO: 1307 LC CDR3 (Combined) QQSFDDPFT SEQ ID NO: 3091 VH QVQLQQSGPELVKPGASVKISCKASGYAFSSSWM NWVKQRPGQGLEWIGRIYPGDGDTKYNGKFKG KATLTADKSSSTAYMHLSSLTSVDSAVYFCARRG TGGWYFDVWGAGTTVTVSS SEQ ID NO: 3284 Heavy chain METDTLLLWVLLLWVPGSTGQVQLQOSGPELVK PGASVKISCKASGYAFSSSWMNWVKQRPGQGLE WIGRIYPGDGDTKYNGKFKGKATLTADKSSSTA YMHLSSLTSVDSAVYFCARRGTGGWYFDVWGA GTTVTVSSAKTTAPSVYPLAPVCGDTTGSSVTLG CLVKGYFPEPVTLTWNSGSLSSGVHTFPAVLQSD LYTLSSSVTVTSSTWPSQSITCNVAHPASSTKVDK KIEPRGPTIKPCPPCKCPAPNLLGGPSVFIFPPKIK DVLMISLSPIVTCVVVDVSEDDPDVQISWFVNNVE VHTAQTQTHREDYNSTLRVVSALPIQHQDWMSG KEFKCKVNNKDLPAPIERTISKPKGSVRAPQVYV LPPPEEEMTKKQVTLTCMVTDFMPEDIYVEWTN NGKTELNYKNTEPVLDSDGSYFMYSKLRVEKKN WVERNSYSCSVVHEGLHNHHTTKSFSRTPGK SEQ ID NO: 3092 VL DIVLTQSPASLAVSLGQRATISCRASESVDSSGNS FMHWYQQKPGQPPQLLIYRASNLESGIPARFSGS GSRTDFTLTINPVEADDVATFYCQQSFDDPFTFGS GTKLEIK SEQ ID NO: 3285 Light chain METDTLLLWVLLLWVPGSTGDIVLTQSPASLAVS LGQRATISCRASESVDSSGNSFMHWYQQKPGQPP QLLIYRASNLESGIPARFSGSGSRTDFTLTINPVEA DDVATFYCQQSFDDPFTFGSGTKLEIKRADAAPT VSIFPPSSEQLTSGGASVVCFLNNFYPKDINVKWK IDGSERQNGVLNSWTDQDSKDSTYSMSSTLTLTK DEYERHNSYTCEATHKTSTSPIVKSFNRNEC Humanized antibody E (E-H antibody) Variable light chain (VL) SEQ ID VL E-H.1 DIVLTQSPDSLAVSLGERATINCRASESVDSSGNSFMHWYQQKPGQ NO: 3093 PPQLLIYRASNLESGVPDRFSGSGSRTDFTLTISSLQAEDVAVYYCQ QSFDDPFTFGQGTKLEIK SEQ ID VL E-H.2 EIVLTQSPATLSLSPGERATLSCRASESVDSSGNSFMHWYQQKPGQ NO: 3094 APQLLIYRASNLESGIPARFSGSGSRTDFTLTISSLEPEDFAVYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.3 EIVLTQSPATLSLSPGERATLSCRASESVDSSGNSFMHWYQQKPGQ NO: 3095 APQLLIYRASNLESGIPARFSGSGSRTDFTLTISRLEPEDFAVYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.4 EIVLTQSPATLSLSPGERATLSCRASESVDSSGNSFMHWYQQKPGQ NO: 3096 APQLLIYRASNLESGIPARFSGSGSRTDFTLTISSLQPEDFAVYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.5 DIQLTQSPSSLSASVGDRVTITCRASESVDSSGNSFMHWYQQKPGQ NO: 3097 APQLLIYRASNLESGVPSRFSGSGSRTDFTLTISSLQPEDVATYYCQ QSFDDPFTFGQGTKLEIK SEQ ID VL E-H.6 EIVLTQSPATLSLSPGERATLSCRASESVDSSGNSFMHWYQQKPGQ NO: 3098 APQLLIYRASNLESGIPARFSGSGPRTDFTLTISSLEPEDFAVYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.7 EIVLTQSPATLSLSPGERATLSCRASESVDSSGNSFMHWYQQKPGQ NO: 3099 APQLLIYRASNLESGIPDRFSGSGSRTDFTLTISRLEPEDFAVYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.8 DIQLTQSPSSLSASVGDRVTITCRASESVDSSGNSFMHWYQQKPGK NO: 3100 VPQLLIYRASNLESGVPSRFSGSGSRTDFTLTISSLQPEDVATYYCQ QSFDDPFTFGQGTKLEIK SEQ ID VL E-H.9 DIQLTQSPSSLSASVGDRVTITCRASESVDSSGNSFMHWYQQKPGK NO: 3101 TPQLLIYRASNLESGIPSRFSGSGSRTDFTLTIRSLQPEDFATYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.10 EIVLTQSPGTLSLSPGERATLSCRASESVDSSGNSFMHWYQQKPGQ NO: 3102 APQLLIYRASNLESGIPDRFSGSGSRTDFTLTISRLEPEDFAVYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.11 EIVLTQSPATLSLSPGERATLSCRASESVDSSGNSFMHWYQQKPGL NO: 3103 APQLLIYRASNLESGIPDRFSGSGSRTDFTLTISRLEPEDFAVYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.12 DIQLTQSPSSLSASVGDRVTITCRASESVDSSGNSFMHWYQQKPGK NO: 3104 APQLLIYRASNLESGVPSRFSGSGSRTDFTLTISSLQPEDFATYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.13 DIQLTQSPSSVSASVGDRVTITCRASESVDSSGNSFMHWYQQKPGK NO: 3105 APQLLIYRASNLESGVPSRFSGSGSRTDFTLTISSLQPEDFATYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.14 AIQLTQSPSSLSASVGDRVTITCRASESVDSSGNSFMHWYQQKPGK NO: 3106 APQLLIYRASNLESGVPSRFSGSGSRTDFTLTISSLQPEDFATYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.15 DIQLTQSPSFLSASVGDRVTITCRASESVDSSGNSFMHWYQQKPGK NO: 3107 APQLLIYRASNLESGVPSRFSGSGSRTEFTLTISSLQPEDFATYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.16 DIQLTQSPSSLSASVGDRVTITCRASESVDSSGNSFMHWYQQKPGK NO: 3108 APQLLIYRASNLESGVPSRFSGSGSRTDFTFTISSLQPEDIATYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.17 EIVLTQSPATLSVSPGERATLSCRASESVDSSGNSFMHWYQQKPGQ NO: 3109 APQLLIYRASNLESGIPARFSGSGSRTEFTLTISILQSEDFAVYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.18 EIVLTQSPATLSVSPGERATLSCRASESVDSSGNSFMHWYQQKPGQ NO: 3110 APQLLIYRASNLESGIPARFSGSGSRTEFTLTISSLQSEDFAVYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.19 AIRLTQSPFSLSASVGDRVTITCRASESVDSSGNSFMHWYQQKPAK NO: 3111 APQLFIYRASNLESGVPSRFSGSGSRTDFTLTISSLQPEDFATYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.20 DIQLTQSPSSLSASVGDRVTITCRASESVDSSGNSFMHWYQQKPGK NO: 3112 APQSLIYRASNLESGVPSRFSGSGSRTDFTLTISSLQPEDFATYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.21 DIQLTQSPSSLSASVGDRVTITCRASESVDSSGNSFMHWYQQKPGK NO: 3113 APQRLIYRASNLESGVPSRFSGSGSRTEFTLTISNLQPEDFATYYCQ QSFDDPFTFGQGTKLEIK SEQ ID VL E-H.22 DIQLTQSPSTLSASVGDRVTITCRASESVDSSGNSFMHWYQQKPGK NO: 3114 APQLLIYRASNLESGVPSRFSGSGSRTEFTLTISSLQPDDFATYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.23 EIVLTQSPDFQSVTPKEKVTITCRASESVDSSGNSFMHWYQQKPDQ NO: 3115 SPQLLIYRASNLESGVPSRFSGSGSRTDFTLTINSLEAEDAATYYCQ QSFDDPFTFGQGTKLEIK SEQ ID VL E-H.24 DIQLTQSPSSLSASVGDRVTITCRASESVDSSGNSFMHWYQQKPGK NO: 3116 APQSLIYRASNLESGVPSKFSGSGSRTDFTLTISSLQPEDFATYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.25 DIQLTQSPSSLSASVGDRVTITCRASESVDSSGNSFMHWYQQKPGK NO: 3117 APQRLIYRASNLESGVPSRFSGSGSRTEFTLTISSLQPEDFATYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.26 DIVLTQTPLSLSVTPGQPASISCRASESVDSSGNSFMHWYLQKPGQP NO: 3118 PQLLIYRASNLESGVPDRFSGSGSRTDFTLKISRVEAEDVGVYYCQ QSFDDPFTFGQGTKLEIK SEQ ID VL E-H.27 DIQLTQSPSSLSASVGDRVTITCRASESVDSSGNSFMHWYQQKPEK NO: 3119 APQSLIYRASNLESGVPSRFSGSGSRTDFTLTISSLQPEDFATYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.28 EIVLTQSPPTLSLSPGERVTLSCRASESVDSSGNSFMHWYQQKPGQ NO: 3120 APQLLIYRASNLESGIPARFSGSGSRTDFTLTISSLQPEDFAVYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.29 DIQLTQSPSAMSASVGDRVTITCRASESVDSSGNSFMHWYQQKPG NO: 3121 KVPQRLIYRASNLESGVPSRFSGSGSRTEFTLTISSLQPEDFATYYCQ QSFDDPFTFGQGTKLEIK SEQ ID VL E-H.30 DIVLTQSPLSLPVTPGEPASISCRASESVDSSGNSFMHWYLQKPGQS NO: 3122 PQLLIYRASNLESGVPDRFSGSGSRTDFTLKISRVEAEDVGVYYCQ QSFDDPFTFGQGTKLEIK SEQ ID VL E-H.31 DIVLTQTPLSLPVTPGEPASISCRASESVDSSGNSFMHWYLQKPGQS NO: 3123 PQLLIYRASNLESGVPDRFSGSGSRTDFTLKISRVEAEDVGVYYCQ QSFDDPFTFGQGTKLEIK SEQ ID VL E-H.32 DIVLTQTPLSLSVTPGQPASISCRASESVDSSGNSFMHWYLQKPGQS NO: 3124 PQLLIYRASNLESGVPDRFSGSGSRTDFTLKISRVEAEDVGVYYCQ QSFDDPFTFGQGTKLEIK SEQ ID VL E-H.33 EIVLTQSPPTLSLSPGERVTLSCRASESVDSSGNSFMHWYQQKPGQ NO: 3125 APQLLIYRASNLESSIPARFSGSGSRTDFTLTISSLQPEDFAVYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.34 DIVLTQSPLSLPVTLGQPASISCRASESVDSSGNSFMHWYQQRPGQS NO: 3126 PQRLIYRASNLESGVPDRFSGSGSRTDFTLKISRVEAEDVGVYYCQ QSFDDPFTFGQGTKLEIK SEQ ID VL E-H.35 DIVLTQTPLSSPVTLGQPASISCRASESVDSSGNSFMHWYQQRPGQP NO: 3127 PQLLIYRASNLESGVPDRFSGSGARTDFTLKISRVEAEDVGVYYCQ QSFDDPFTFGQGTKLEIK SEQ ID VL E-H.36 DIVLTQSPAFLSVTPGEKVTITCRASESVDSSGNSFMHWYQQKPDQ NO: 3128 APQLLIYRASNLESGVPSRFSGSGSRTDFTFTISSLEAEDAATYYCQ QSFDDPFTFGQGTKLEIK SEQ ID VL E-H.37 DIQLIQSPSFLSASVGDRVSIICRASESVDSSGNSFMHWYLQKPGKS NO: 3129 PQLFIYRASNLESGVSSRFSGRGSRTDFTLTIISLKPEDFAAYYCQQS FDDPFTFGQGTKLEIK SEQ ID VL E-H.38 EIVLTQTPLSLSITPGEQASISCRASESVDSSGNSFMHWYLQKARPV NO: 3130 PQLLIYRASNLESGVPDRFSGSGSRTDFTLKISRVEAEDFGVYYCQQ SFDDPFTFGQGTKLEIK SEQ ID VL E-H.39 EIVLTQTPLSLSITPGEQASMSCRASESVDSSGNSFMHWYLQKARP NO: 3131 VPQLLIYRASNLESGVPDRFSGSGSRTDFTLKISRVEAEDFGVYYCQ QSFDDPFTFGQGTKLEIK SEQ ID VL E-H.40 EITLTQSPAFMSATPGDKVNISCRASESVDSSGNSFMHWYQQKPGE NO: 3132 APQFIIYRASNLESGIPPRFSGSGYRTDFTLTINNIESEDAAYYYCQQ SFDDPFTFGQGTKLEIK Variable HEAVY chain (VH) SEQ ID VH E-H.1 QVQLVQSGAEVKKPGASVKVSCKASGYAFSSSWMNWVRQAPGQ NO: 3133 GLEWIGRIYPGDGDTKYNGKFKGRATLTADKSTSTAYMELSSLRS EDTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.2 QVQLVQSGAEVKKPGSSVKVSCKASGYAFSSSWMNWVRQAPGQ NO: 3134 GLEWIGRIYPGDGDTKYNGKFKGRATLTADKSTSTAYMELSSLRS EDTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.3 QVQLVQSGAEVKKPGASVKVSCKASGYAFSSSWMNWVRQAPGK NO: 3135 GLEWIGRIYPGDGDTKYNGKFKGRATLTADKSTSTAYMELSSLRS EDTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.4 QVQLVQSGAEVKKPGASVKVSCKASGYAFSSSWMNWVRQAPGQ NO: 3136 ELEWIGRIYPGDGDTKYNGKFKGRATLTADKSISTAYMELSSLRSE DTATYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.5 EVQLVQSGAEVKKPGATVKISCKASGYAFSSSWMNWVQQAPGKG NO: 3137 LEWIGRIYPGDGDTKYNGKFKGRATLTADKSTSTAYMELSSLRSE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.6 QVQLVQSGAEVKKTGSSVKVSCKASGYAFSSSWMNWVRQAPGQ NO: 3138 ALEWIGRIYPGDGDTKYNGKFKGRATLTADKSMSTAYMELSSLRS EDTAMYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.7 QVQLVQSGAEVKKPGASVKVSCKASGYAFSSSWMNWVRQAPGQ NO: 3139 RLEWIGRIYPGDGDTKYNGKFKGRATLTADKSASTAYMELSSLRS EDMAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.8 QVQLVQSGAEVKKPGASVKVSCKASGYAFSSSWMNWVRQAPGQ NO: 3140 GLEWIGRIYPGDGDTKYNGKFKGRATLTADKSTSTAYMELRSLRS DDMAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.9 QVQLVQSGAEVKKPGASVKVSCKASGYAFSSSWMNWVRQAPGQ NO: 3141 RLEWIGRIYPGDGDTKYNGKFKGRATLTADKSASTAYMELSSLRS EDTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.10 QVQLVQSGAEVKKPGASVKVSCKASGYAFSSSWMNWVRQAPGQ NO: 3142 GLEWIGRIYPGDGDTKYNGKFKGRATLTADKSTSTAYMELRSLRS DDTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.11 QVQLVQSGAEVKKPGASVKVSCKASGYAFSSSWMNWVRQAPGQ NO: 3143 GLEWIGRIYPGDGDTKYNGKFKGRATLTADKSISTAYMELSRLRSD DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.12 QVQLVQSGAEVKKPGASVKVSCKASGYAFSSSWMNWVRQAPGQ NO: 3144 GLEWIGRIYPGDGDTKYNGKFKGRATLTADKSISTAYMELSRLRSD DTVVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.13 QVQLVQSGAEVKKPGASVKVSCKASGYAFSSSWMNWVRQAPGQ NO: 3145 GLEWIGRIYPGDGDTKYNGKFKGWATLTADKSISTAYMELSRLRS DDTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.14 QVQLVQSGAEVKKPGASVKVSCKASGYAFSSSWMNWVRQATGQ NO: 3146 GLEWIGRIYPGDGDTKYNGKFKGRATLTANKSISTAYMELSSLRSE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.15 QVQLVQSGSELKKPGASVKVSCKASGYAFSSSWMNWVRQAPGQG NO: 3147 LEWIGRIYPGDGDTKYNGKFKGRAVLSADKSVSTAYLQISSLKAED TAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.16 QVQLVQSGPEVKKPGTSVKVSCKASGYAFSSSWMNWVRQARGQ NO: 3148 RLEWIGRIYPGDGDTKYNGKFKGRATLTADKSTSTAYMELSSLRSE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.17 EVQLVQSGAEVKKPGESLKISCKASGYAFSSSWMNWVRQMPGKG NO: 3149 LEWIGRIYPGDGDTKYNGKFKGQATLSADKSISTAYLQWSSLKAS DTAMYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.18 QVQLVQSGSELKKPGASVKVSCKASGYAFSSSWMNWVRQAPGQG NO: 3150 LEWIGRIYPGDGDTKYNGKFKGRAVLSADKSVSMAYLQISSLKAE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.19 QVQLVQSGHEVKQPGASVKVSCKASGYAFSSSWMNWVPQAPGQ NO: 3151 GLEWIGRIYPGDGDTKYNGKFKGRAVLSADKSASTAYLQISSLKAE DMAMYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.20 EVQLVQSGAEVKKPGESLKISCKASGYAFSSSWMNWVRQMPGKG NO: 3152 LEWIGRIYPGDGDTKYNGKFKGQATLSADKPISTAYLQWSSLKAS DTAMYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.21 EVQLVQSGAEVKKPGESLRISCKASGYAFSSSWMNWVRQMPGKG NO: 3153 LEWIGRIYPGDGDTKYNGKFKGQATLSADKSISTAYLQWSSLKAS DTAMYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.22 EVQLVQSGAEVKKPGESLRISCKASGYAFSSSWMNWVRQMPGKG NO: 3154 LEWIGRIYPGDGDTKYNGKFKGHATLSADKSISTAYLQWSSLKAS DTAMYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.23 QVQLVQSGAEVKKTGSSVKVSCKASGYAFSSSWMNWVRQAPRQ NO: 3155 ALEWIGRIYPGDGDTKYNGKFKGRATLTADKSMSTAYMELSSLRS EDTAMYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.24 EVQLVESGGGLVQPGRSLRLSCTASGYAFSSSWMNWVRQAPGKG NO: 3156 LEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSIAYLQMNSLKTE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.25 EVQLVESGGGLVQPGPSLRLSCTASGYAFSSSWMNWVRQAPGKG NO: 3157 LEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSIAYLQMNSLKTE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.26 QVQLQESGPGLVKPSQTLSLTCTASGYAFSSSWMNWVRQPPGKGL NO: 3158 EWIGRIYPGDGDTKYNGKFKGRATLSADKSKSQASLKLSSVTAAD TAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.27 QVQLQESGPGLVKPSGTLSLTCAASGYAFSSSWMNWVRQPPGKGL NO: 3159 EWIGRIYPGDGDTKYNGKFKGRATLSADKSKSQASLKLSSVTAAD TAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.28 EVQLVESGGGLVKPGRSLRLSCTASGYAFSSSWMNWVRQAPGKG NO: 3160 LEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSIAYLQMNSLKTE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.29 EVQLVESGGGLVQPGGSLKLSCAASGYAFSSSWMNWVRQASGKG NO: 3161 LEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSTAYLQMNSLKTE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.30 QVQLQESGPGLVKPSQTLSLTCAASGYAFSSSWMNWVRQPPGKGL NO: 3162 EWIGRIYPGDGDTKYNGKFKGRATLSADKSKSQASLKLSSVTAAD TAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.31 EVQLVESGGGLVKPGGSLRLSCAASGYAFSSSWMNWVRQAPGKG NO: 3163 LEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSTAYLQMNSLKTE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.32 EVQLVESGGALVKPGGSLRLSCAASGYAFSSSWMNWVRQAPGKG NO: 3164 LEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSTAYLQMNSLKTE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.33 QVQLQESGPGLVKPSQTLSLTCAAYGYAFSSSWMNWVRQPPGKG NO: 3165 LEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSQASLKLSSVTAA DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.34 QVQLQESGSGLVKPSQTLSLTCAASGYAFSSSWMNWVRQPPGKGL NO: 3166 EWIGRIYPGDGDTKYNGKFKGRATLSADKSKSQASLKLSSVTAAD TAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.35 EVQLVESGGGLVQPGGSLRLSCAASGYAFSSSWMNWVRQAPGKG NO: 3167 LEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSSAYLQMNSLKTE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.36 QVQLQESGPGLVKPSDTLSLTCTASGYAFSSSWMNWVRQPPGKGL NO: 3168 EWIGRIYPGDGDTKYNGKFKGRATLSADKSKSQASLKLSSVTAAD TAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.37 QVQLQESGPGLVKPSQTLSLTCTASGYAFSSSWMNWVRQHPGKG NO: 3169 LEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSQASLKLSSVTAA DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.38 QVQLQESGPGLVKPSQTLSLTCTASGYAFSSSWMNWVRQHPGKG NO: 3170 LEWIGRIYPGDGDTKYNGKFKGLATLSADKSKSQASLKLSSVTAA DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.39 QVQLVESGGGVVQPGRSLRLSCAASGYAFSSSWMNWVRQAPGKG NO: 3171 LEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSTAYLQMSSLRAE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.40 QVQLVESGGGLVKPGGSLRLSCAASGYAFSSSWMNWVRQAPGKG NO: 3172 LEWIGRIYPGDGDTKYNGKFKGRATLSADKAKSSAYLQMNSLRAE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.41 QVQLVESGGGLVQPGGSLRLSCSASGYAFSSSWMNWVRQAPGKG NO: 3173 LEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSTAYLQMNSLRAE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.42 QVQLLESGGGLVKPGGSLRLSCAASGYAFSSSWMNWVRQAPGKG NO: 3174 LEWIGRIYPGDGDTKYNGKFKGRATLSADKAKSSAYLQMNSLRAE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.43 EVQLVESGGGLVQPGGSLRLSCSASGYAFSSSWMNWVRQAPGKG NO: 3175 LEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSTAYLQMSSLRAE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.44 QVQLQESGPGLVKPSDTLSLTCAASGYAFSSSWMNWVRQPPGKGL NO: 3176 EWIGRIYPGDGDTKYNGKFKGRATLSADKSKSQASLKLSSVTAVD TAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.45 QVQLQESGPGLVKPSQTLSLTCAASGYAFSSSWMNWVRQPPGKGL NO: 3177 EWIGRIYPGDGDTKYNGKFKGRATLSADKSKSQASLKLSSVTAVD TAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.46 EVQLVESGGGLVQPGGSLRLSCSASGYAFSSSWMNWVRQAPGKG NO: 3178 LEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSTAYVQMSSLRAE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.47 QVQLVDSGGGVVQPGRSLRLSCAASGYAFSSSWMNWVRQAPGK NO: 3179 GLEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSTAYLQMNSLRA EDTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.48 QVQLVESGGGVVQPGRSLRLSCAASGYAFSSSWMNWVRQAPGKG NO: 3180 LEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSTAYLQMNSLRAE GTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.49 QVQLVESGGGVVQPGRSLRLSCAASGYAFSSSWMNWVRQAPGKG NO: 3181 LEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSTAYLQMNSLRAE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS SEQ ID VH E-H.50 EVQLVESGGGLVQPGGSLRLSCAASGYAFSSSWMNWVRQAPGKG NO: 3182 LEWIGRIYPGDGDTKYNGKFKGRATLSADKSKSTAYLQMNSLRAE DTAVYYCARRGTGGWYFDVWGQGTTVTVSS

In some embodiments, the anti-TCRβ V5 antibody molecule comprises a VH and/or a VL of an antibody described in Table 10, or a sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.

In some embodiments, the anti-TCRβ V5 antibody molecule comprises a VH and a VL of an antibody described in Table 10, or a sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.

In some embodiments, the anti-TCRβ V5 antibody molecule comprises a VH and/or a VL of an antibody described in Table 11, or a sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.

In some embodiments, the anti-TCRβ V5 antibody molecule comprises a VH and a VL of an antibody described in Table 11, or a sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.

Anti-TCRβ V10 Antibodies

Accordingly, in one aspect, the disclosure provides an anti-TCRβV antibody molecule that binds to a human TCRβ V10 subfamily member. In some embodiments, TCRβ V10 subfamily is also known as TCRβ V12. In some embodiments, the TCRβ V10 subfamily comprises: TCRβ V10-1*01, TCRβ V10-1*02, TCRβ V10-3*01 or TCRβ V10-2*01, or a variant thereof.

Exemplary anti-TCRβ V10 antibodies of the disclosure are provided in Table 12. In some embodiments, the anti-TCRβ V10 is antibody D, e.g., humanized antibody D (antibody D-H), as provided in Table 12. In some embodiments, antibody D comprises one or more (e.g., three) light chain CDRs and/or one or more (e.g., three) heavy chain CDRs provided in Table 12, or a sequence with at least 95% identity thereto. In some embodiments, antibody D comprises a variable heavy chain (VH) and/or a variable light chain (VL) provided in Table 12, or a sequence with at least 95% identity thereto.

TABLE 12 Amino acid sequences for anti TCRβ V10 antibodies Amino acid and nucleotide sequences for murine and humanized antibody molecules which bind to TCRBV 10 (e.g., TCRBV 10-1, TCRBV 10-2 or TCRBV 10-3). The amino acid the heavy and light chain CDRs, and the amino acid and nucleotide sequences of the heavy and light chain variable regions, and the heavy and light chains are shown. Murine antibody D, also referred to as S511 antibody SEQ ID NO: 1288 HC CDR1 (Kabat) SYGMS SEQ ID NO: 1289 HC CDR2 (Kabat) LISSGGSYTYYTDSVKG SEQ ID NO: 1290 HC CDR3 (Kabat) HGGNFFDY SEQ ID NO: 1291 HC CDR1 (Chothia) GFTFRSY SEQ ID NO: 1292 HC CDR2 (Chothia) SSGGSY SEQ ID NO: 1290 HC CDR3 (Chothia) HGGNFFDY SEQ ID NO: 1293 HC CDR1 (Combined) GFTFRSYGMS SEQ ID NO: 1289 HC CDR2 (Combined) LISSGGSYTYYTDSVKG SEQ ID NO: 1290 HC CDR3 (Combined) HGGNFFDY SEQ ID NO: 1294 LC CDR1 (Kabat) SVSSSVSYMH SEQ ID NO: 1295 LC CDR2 (Kabat) DTSKLAS SEQ ID NO: 1296 LC CDR3 (Kabat) QQWSSNPQYT SEQ ID NO: 1297 LC CDR1 (Chothia) SSSVSY SEQ ID NO: 1295 LC CDR2 (Chothia) DTSKLAS SEQ ID NO: 1296 LC CDR3 (Chothia) QQWSSNPQYT SEQ ID NO: 1294 LC CDR1 (Combined) SVSSSVSYMH SEQ ID NO: 1295 LC CDR2 (Combined) DTSKLAS SEQ ID NO: 1296 LC CDR3 (Combined) QQWSSNPQYT SEQ ID NO: 3183 VH EVQLVESGGDLVKPGGSLKLSCAVSGFTFRSYGM SWVRQTPDKRLEWVALISSGGSYTYYTDSVKGR FTISRDNAKNTLYLQMSSLKSEDTAIYYCSRHGG NFFDYWGQGTTLTVSS SEQ ID NO: 3184 VL QIVLTQSPSIMSASPGEKVTMTCSVSSSVSYMHW YQQKSGTSPKRWIYDTSKLASGVPARFSGSGSGT SYSLTISSMEAEDAATYYCQQWSSNPQYTFGGGT KLEIK Humanized antibody D (D-H antibody) Variable light chain (VL) SEQ ID VL D-H.1 DIVLTQSPAFLSVTPGEKVTITCSVSSSVSYMHWYQQKPDQAPKLLI NO: 3185 YDTSKLASGVPSRFSGSGSGTDYTFTISSLEAEDAATYYCQQWSSN PQYTFGQGTKLEIK SEQ ID VL D-H.2 AIQLTQSPSSLSASVGDRVTITCSVSSSVSYMHWYQQKPGKAPKLLI NO: 3186 YDTSKLASGVPSRFSGSGSGTDYTLTISSLQPEDFATYYCQQWSSN PQYTFGQGTKLEIK SEQ ID VL D-H.3 DIQLTQSPSFLSASVGDRVTITCSVSSSVSYMHWYQQKPGKAPKLLI NO: 3187 YDTSKLASGVPSRFSGSGSGTEYTLTISSLQPEDFATYYCQQWSSNP QYTFGQGTKLEIK SEQ ID VL D-H.4 DIQLTQSPSSLSASVGDRVTITCSVSSSVSYMHWYQQKPGKAPKLLI NO: 3188 YDTSKLASGVPSRFSGSGSGTDYTLTISSLQPEDFATYYCQQWSSN PQYTFGQGTKLEIK SEQ ID VL D-H.5 DIQLTQSPSSVSASVGDRVTITCSVSSSVSYMHWYQQKPGKAPKLL NO: 3189 IYDTSKLASGVPSRFSGSGSGTDYTLTISSLQPEDFATYYCQQWSSN PQYTFGQGTKLEIK SEQ ID VL D-H.6 DIQLTQSPSSLSASVGDRVTITCSVSSSVSYMHWYQQKPGKVPKLLI NO: 3190 YDTSKLASGVPSRFSGSGSGTDYTLTISSLQPEDVATYYCQQWSSN PQYTFGQGTKLEIK SEQ ID VL D-H.7 DIQLTQSPSSLSASVGDRVTITCSVSSSVSYMHWYQQKPGQAPKLLI NO: 3191 YDTSKLASGVPSRFSGSGSGTDYTLTISSLQPEDVATYYCQQWSSN PQYTFGQGTKLEIK SEQ ID VL D-H.8 EIVLTQSPDFQSVTPKEKVTITCSVSSSVSYMHWYQQKPDQSPKLLI NO: 3192 YDTSKLASGVPSRFSGSGSGTDYTLTINSLEAEDAATYYCQQWSSN PQYTFGQGTKLEIK SEQ ID VL D-H.9 AIRLTQSPFSLSASVGDRVTITCSVSSSVSYMHWYQQKPAKAPKLFI NO: 3193 YDTSKLASGVPSRFSGSGSGTDYTLTISSLQPEDFATYYCQQWSSN PQYTFGQGTKLEIK SEQ ID VL D-H.10 DIQLTQSPSSLSASVGDRVTITCSVSSSVSYMHWYQQKPGKAPKLLI NO: 3194 YDTSKLASGVPSRFSGSGSGTDYTFTISSLQPEDIATYYCQQWSSNP QYTFGQGTKLEIK SEQ ID VL D-H.11 EIVLTQSPATLSLSPGERATLSCSVSSSVSYMHWYQQKPGQAPKLLI NO: 3195 YDTSKLASGIPARFSGSGSGTDYTLTISSLEPEDFAVYYCQQWSSNP QYTFGQGTKLEIK SEQ ID VL D-H.12 DIQLTQSPSTLSASVGDRVTITCSVSSSVSYMHWYQQKPGKAPKLL NO: 3196 IYDTSKLASGVPSRFSGSGSGTEYTLTISSLQPDDFATYYCQQWSSN PQYTFGQGTKLEIK SEQ ID VL D-H.13 DIQLTQSPSSLSASVGDRVTITCSVSSSVSYMHWYQQKPGKTPKLLI NO: 3197 YDTSKLASGIPSRFSGSGSGTDYTLTIRSLQPEDFATYYCQQWSSNP QYTFGQGTKLEIK SEQ ID VL D-H.14 EIVLTQSPPTLSLSPGERVTLSCSVSSSVSYMHWYQQKPGQAPKLLI NO: 3198 YDTSKLASGIPARFSGSGSGTDYTLTISSLQPEDFAVYYCQQWSSNP QYTFGQGTKLEIK SEQ ID VL D-H.15 DIQLTQSPSSLSASVGDRVTITCSVSSSVSYMHWYQQKPGKAPKRL NO: 3199 IYDTSKLASGVPSRFSGSGSGTEYTLTISSLQPEDFATYYCQQWSSN PQYTFGQGTKLEIK SEQ ID VL D-H.16 EIVLTQSPATLSLSPGERATLSCSVSSSVSYMHWYQQKPGQAPKLLI NO: 3200 YDTSKLASGIPARFSGSGPGTDYTLTISSLEPEDFAVYYCQQWSSNP QYTFGQGTKLEIK SEQ ID VL D-H.17 EIVLTQSPATLSLSPGERATLSCSVSSSVSYMHWYQQKPGQAPKLLI NO: 3201 YDTSKLASGIPARFSGSGSGTDYTLTISRLEPEDFAVYYCQQWSSNP QYTFGQGTKLEIK SEQ ID VL D-H.18 EIVLTQSPATLSLSPGERATLSCSVSSSVSYMHWYQQKPGQAPKLLI NO: 3202 YDTSKLASGIPARFSGSGSGTDYTLTISSLQPEDFAVYYCQQWSSNP QYTFGQGTKLEIK SEQ ID VL D-H.19 EIVLTQSPATLSVSPGERATLSCSVSSSVSYMHWYQQKPGQAPKLL NO: 3203 IYDTSKLASGIPARFSGSGSGTEYTLTISSLQSEDFAVYYCQQWSSN PQYTFGQGTKLEIK SEQ ID VL D-H.20 EIVLTQSPATLSVSPGERATLSCSVSSSVSYMHWYQQKPGQAPKLL NO: 3204 IYDTSKLASGIPARFSGSGSGTEYTLTISILQSEDFAVYYCQQWSSN PQYTFGQGTKLEIK SEQ ID VL D-H.21 EIVLTQSPPTLSLSPGERVTLSCSVSSSVSYMHWYQQKPGQAPKLLI NO: 3205 YDTSKLASSIPARFSGSGSGTDYTLTISSLQPEDFAVYYCQQWSSNP QYTFGQGTKLEIK SEQ ID VL D-H.22 DIQLTQSPSSLSASVGDRVTITCSVSSSVSYMHWYQQKPGKAPKSLI NO: 3206 YDTSKLASGVPSRFSGSGSGTDYTLTISSLQPEDFATYYCQQWSSN PQYTFGQGTKLEIK SEQ ID VL D-H.23 DIQLTQSPSSLSASVGDRVTITCSVSSSVSYMHWYQQKPGKAPKRL NO: 3207 IYDTSKLASGVPSRFSGSGSGTEYTLTISNLQPEDFATYYCQQWSSN PQYTFGQGTKLEIK SEQ ID VL D-H.24 DIQLTQSPSAMSASVGDRVTITCSVSSSVSYMHWYQQKPGKVPKR NO: 3208 LIYDTSKLASGVPSRFSGSGSGTEYTLTISSLQPEDFATYYCQQWSS NPQYTFGQGTKLEIK SEQ ID VL D-H.25 EIVLTQSPATLSLSPGERATLSCSVSSSVSYMHWYQQKPGQAPKLLI NO: 3209 YDTSKLASGIPDRFSGSGSGTDYTLTISRLEPEDFAVYYCQQWSSNP QYTFGQGTKLEIK SEQ ID VL D-H.26 EIVLTQSPATLSLSPGERATLSCSVSSSVSYMHWYQQKPGLAPKLLI NO: 3210 YDTSKLASGIPDRFSGSGSGTDYTLTISRLEPEDFAVYYCQQWSSNP QYTFGQGTKLEIK SEQ ID VL D-H.27 EIVLTQSPGTLSLSPGERATLSCSVSSSVSYMHWYQQKPGQAPKLLI NO: 3211 YDTSKLASGIPDRFSGSGSGTDYTLTISRLEPEDFAVYYCQQWSSNP QYTFGQGTKLEIK SEQ ID VL D-H.28 DIQLTQSPSSLSASVGDRVTITCSVSSSVSYMHWYQQKPGKAPKSLI NO: 3212 YDTSKLASGVPSKFSGSGSGTDYTLTISSLQPEDFATYYCQQWSSN PQYTFGQGTKLEIK SEQ ID VL D-H.29 DIQLTQSPSSLSASVGDRVTITCSVSSSVSYMHWYQQKPEKAPKSLI NO: 3213 YDTSKLASGVPSRFSGSGSGTDYTLTISSLQPEDFATYYCQQWSSN PQYTFGQGTKLEIK SEQ ID VL D-H.30 DIVLTQSPDSLAVSLGERATINCSVSSSVSYMHWYQQKPGQPPKLL NO: 3214 IYDTSKLASGVPDRFSGSGSGTDYTLTISSLQAEDVAVYYCQQWSS NPQYTFGQGTKLEIK SEQ ID VL D-H.31 EIVLTQTPLSLSITPGEQASMSCSVSSSVSYMHWYLQKARPVPKLLI NO: 3215 YDTSKLASGVPDRFSGSGSGTDYTLKISRVEAEDFGVYYCQQWSS NPQYTFGQGTKLEIK SEQ ID VL D-H.32 EIVLTQTPLSLSITPGEQASISCSVSSSVSYMHWYLQKARPVPKLLIY NO: 3216 DTSKLASGVPDRFSGSGSGTDYTLKISRVEAEDFGVYYCQQWSSNP QYTFGQGTKLEIK SEQ ID VL D-H.33 DIVLTQSPLSLPVTPGEPASISCSVSSSVSYMHWYLQKPGQSPKLLI NO: 3217 YDTSKLASGVPDRFSGSGSGTDYTLKISRVEAEDVGVYYCQQWSS NPQYTFGQGTKLEIK SEQ ID VL D-H.34 DIVLTQSPLSLPVTLGQPASISCSVSSSVSYMHWYQQRPGQSPKRLI NO: 3218 YDTSKLASGVPDRFSGSGSGTDYTLKISRVEAEDVGVYYCQQWSS NPQYTFGQGTKLEIK SEQ ID VL D-H.35 DIVLTQTPLSLPVTPGEPASISCSVSSSVSYMHWYLQKPGQSPKLLI NO: 3219 YDTSKLASGVPDRFSGSGSGTDYTLKISRVEAEDVGVYYCQQWSS NPQYTFGQGTKLEIK SEQ ID VL D-H.36 DIVLTQTPLSLSVTPGQPASISCSVSSSVSYMHWYLQKPGQSPKLLI NO: 3220 YDTSKLASGVPDRFSGSGSGTDYTLKISRVEAEDVGVYYCQQWSS NPQYTFGQGTKLEIK SEQ ID VL D-H.37 DIVLTQTPLSLSVTPGQPASISCSVSSSVSYMHWYLQKPGQPPKLLI NO: 3221 YDTSKLASGVPDRFSGSGSGTDYTLKISRVEAEDVGVYYCQQWSS NPQYTFGQGTKLEIK SEQ ID VL D-H.38 DIQLIQSPSFLSASVGDRVSIICSVSSSVSYMHWYLQKPGKSPKLFIY NO: 3222 DTSKLASGVSSRFSGRGSGTDYTLTIISLKPEDFAAYYCQQWSSNP QYTFGQGTKLEIK SEQ ID VL D-H.39 DIVLTQTPLSSPVTLGQPASISCSVSSSVSYMHWYQQRPGQPPKLLI NO: 3223 YDTSKLASGVPDRFSGSGAGTDYTLKISRVEAEDVGVYYCQQWSS NPQYTFGQGTKLEIK SEQ ID VL D-H.40 EITLTQSPAFMSATPGDKVNISCSVSSSVSYMHWYQQKPGEAPKFII NO: 3224 YDTSKLASGIPPRFSGSGYGTDYTLTINNIESEDAAYYYCQQWSSN PQYTFGQGTKLEIK Variable HEAVY chain (VH) SEQ ID VH D-H.1 EVQLVESGGGLVKPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3225 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMNSLKTEDT AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.2 EVQLVESGGALVKPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3226 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMNSLKTEDT AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.3 EVQLVESGGGLVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3227 EWVALISSGGSYTYYTDSVKGRFTISRDNAKNTLYLQMNSLRAED TAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.4 EVQLVESGGGLVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3228 EWVALISSGGSYTYYTDSVKGRFTISRDNAKNSLYLQMNSLRAED TAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.5 EVQLVESGGGLVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3229 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNSLYLQMNSLKTEDT AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.6 EVQLVESGGGLVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3230 EWVALISSGGSYTYYTDSVKGRFTISRDNAKNSLYLQMNSLRAED MAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.7 EVQLVESGGGLVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3231 EWVALISSGGSYTYYTDSVKGQFTISRDNAKNTLYLQMNSLRAED MAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.8 EVQLVESGGGLVKPGRSLRLSCTVSGFTFRSYGMSWVRQAPGKGL NO: 3232 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNILYLQMNSLKTEDT AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.9 EVQLVESGGGLVKPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3233 EWVALISSGGSYTYYTDSVKGRFTISRDNAKNSLYLQMNSLRAED TAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.10 EVQLVESGGGLVQPGGSLKLSCAVSGFTFRSYGMSWVRQASGKG NO: 3234 LEWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMNSLKTED TAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.11 QVQLVESGGGVVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKG NO: 3235 LEWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMNSLRAED TAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.12 QVQLVESGGGVVQPGRSLRLSCAVSGFTFRSYGMSWVRQAPGKG NO: 3236 LEWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMSSLRAED TAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.13 EVQLVESGGGLVQPGGSLRLSCPVSGFTFRSYGMSWVRQAPGKGL NO: 3237 EWVALISSGGSYTYYTDSVKGRFTISRDNANNSLYLQMNSLRAED TAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.14 EVQLVESGGGLVQPGRSLRLSCTVSGFTFRSYGMSWVRQAPGKGL NO: 3238 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNILYLQMNSLKTEDT AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.15 EVQLVESGGGLVQPGPSLRLSCTVSGFTFRSYGMSWVRQAPGKGL NO: 3239 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNILYLQMNSLKTEDT AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.16 EVQLVESGGGLVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3240 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMNSLRAEDT AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.17 EVQLVESGGGLVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3241 EWVALISSGGSYTYYTDSVKGRFTISRDNAKNSLYLQMNSLRDED TAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.18 QVQLVESGGGLVKPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKG NO: 3242 LEWVALISSGGSYTYYTDSVKGRFTISRDNAKNSLYLQMNSLRAE DTAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.19 QVQLVESGGGVVQPGRSLRLSCAVSGFTFRSYGMSWVRQAPGKG NO: 3243 LEWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMNSLRAED TAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.20 EVQLLESGGGLVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3244 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMNSLRAEDT AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.21 EVQLVESGGGLVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3245 EWVALISSGGSYTYYTDSVKGRFTISRHNSKNTLYLQMNSLRAEDT AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.22 EVQLVESGGGLIQPGGSLRLSCAVSGFTFRSYGMSWVRQPPGKGL NO: 3246 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMNSLRAEDT AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.23 EVQLVESGGGLIQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3247 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMNSLRAEDT AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.24 EVQLVESGGGLVQPGRSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3248 EWVALISSGGSYTYYTDSVKGRFTISRDNAKNSLYLQMNSLRAED TALYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.25 QVQLVESGGGVVQPGRSLRLSCAVSGFTFRSYGMSWVRQAPGKG NO: 3249 LEWVALISSGGSYTYYTDSVKGRFTISRDNSKNRLYLQMNSLRAE DTAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.26 QVQLVESGGGVVQPGRSLRLSCAVSGFTFRSYGMSWVRQAPGKG NO: 3250 LEWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMNSLRAEG TAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.27 QVQLVESGGGVVQPGRSLRLSCAVSGFTFRSYGMSWVRQAPGKG NO: 3251 LEWVALISSGGSYTYYTDSVKGRFAISRDNSKNTLYLQMNSLRAE DTAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.28 QVQLVDSGGGVVQPGRSLRLSCAVSGFTFRSYGMSWVRQAPGKG NO: 3252 LEWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMNSLRAED TAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.29 EVQLVESGGGVVRPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKG NO: 3253 LEWVALISSGGSYTYYTDSVKGRFTISRDNAKNSLYLQMNSLRAE DTALYHCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.30 EVQLVESGGVVVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKG NO: 3254 LEWVALISSGGSYTYYTDSVKGRFTISRDNSKNSLYLQMNSLRAED TALYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.31 EVQLVESGGGVVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKG NO: 3255 LEWVALISSGGSYTYYTDSVKGRFTISRDNSKNSLYLQMNSLRTED TALYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.32 EVQLVESGGVVVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKG NO: 3256 LEWVALISSGGSYTYYTDSVKGRFTISRDNSKNSLYLQMNSLRTED TALYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.33 EVQLVETGGGLIQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3257 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMNSLRAEDT AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.34 EVQLVESGGGLVQPGGSLRLSCAVSGFTFRSYGMSWVRQATGKG NO: 3258 LEWVALISSGGSYTYYTDSVKGRFTISRENAKNSLYLQMNSLRAG DTAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.35 EVQLVESRGVLVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3259 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLHLQMNSLRAEDT AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.36 EVQLVESGGGLVQPGRSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3260 EWVALISSGGSYTYYTDSVKGRFTISRDNAKNSLYLQMNSLRAED MALYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.37 QVQLVESGGGLVQPGGSLRLSCSVSGFTFRSYGMSWVRQAPGKGL NO: 3261 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMNSLRAEDT AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.38 EVQLVESGGGLVQPGGSLRLSCSVSGFTFRSYGMSWVRQAPGKGL NO: 3262 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMSSLRAEDT AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.39 QVQLVESGGGVVQPGRSLRLSCAVSGFTFRSYGMSWVRQAPGKG NO: 3263 LEWVALISSGGSYTYYTDSVKGRFTISRDNSTNTLFLQMNSLRAED TAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.40 QVQLLESGGGLVKPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3264 EWVALISSGGSYTYYTDSVKGRFTISRDNAKNSLYLQMNSLRAED TAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.41 EVQLVESGEGLVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3265 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMGSLRAED MAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.42 EVQLVESGGGLVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3266 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMGSLRAED MAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.43 EVQLVESGGGLVQPGGSLRLSCSVSGFTFRSYGMSWVRQAPGKGL NO: 3267 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYVQMSSLRAEDT AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.44 EVQLVESGGGLVQPGGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3268 EWVALISSGGSYTYYTDSVKGRFIISRDNSRNSLYLQKNRRRAEDM AVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.45 EVQLVESGGGLVQPGGSLRLSCAVSGFTFRSYGMSWVHQAPGKG NO: 3269 LEWVALISSGGSYTYYTDSVKGRFIISRDNSRNTLYLQTNSLRAE DTAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.46 EVHLVESGGGLVQPGGALRLSCAVSGFTFRSYGMSWVRQATGKG NO: 3270 LEWVALISSGGSYTYYTDSVKGRFTISRENAKNSLYLQMNSLRAG DTAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.47 EVQLVESGGGLVQPRGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3271 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMNNLRAEG TAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.48 EVQLVESGGGLVQPRGSLRLSCAVSGFTFRSYGMSWVRQAPGKGL NO: 3272 EWVALISSGGSYTYYTDSVKGRFTISRDNSKNTLYLQMNNLRAEG TAAYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.49 QVQLVQSGAEVKKPGASVKVSCKVSGFTFRSYGMSWVRQAPGKG NO: 3273 LEWVALISSGGSYTYYTDSVKGRFTITRDNSTNTLYMELSSLRSED TAVYYCSRHGGNFFDYWGQGTTVTVSS SEQ ID VH D-H.50 QVQLVQSGSELKKPGASVKVSCKVSGFTFRSYGMSWVRQAPGQG NO: 3274 LEWVALISSGGSYTYYTDSVKGRFVISRDNSVNTLYLQISSLKAE DTAVYYCSRHGGNFFDYWGQGTTVTVSS

In some embodiments, the anti-TCRβ V10 antibody molecule comprises a VH or a VL of an antibody described in Table 12, or a sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.

In some embodiments, the anti-TCRβ V10 antibody molecule comprises a VH and a VL of an antibody described in Table 12, or a sequence with at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or more identity thereto.

Additional Anti-TCRVβ Antibodies

Additional exemplary anti-TCRβV antibodies of the disclosure are provided in Table 13. In some embodiments, the anti-TCRβV antibody is a humanized antibody, e.g., as provided in Table 13. In some embodiments, the anti-TCRβV antibody comprises one or more (e.g., all three) of a LC CDR1, LC CDR2, and LC CDR3 provided in Table 13; and/or one or more (e.g., all three) of a HC CDR1, HC CDR2, and HC CDR3 provided in Table 13, or a sequence with at least 95% identity thereto. In some embodiments, the anti-TCRβV antibody comprises a variable heavy chain (VH) and/or a variable light chain (VL) provided in Table 13, or a sequence with at least 95% identity thereto.

TABLE 13 Amino acid sequences for additional anti-TCRβ V antibodies Amino acid and nucleotide sequences for murine and humanized antibody molecules which bind to various TCRVB families are disclosed. The amino acid the heavy and light chain CDRs, and the amino acid and nucleotide sequences of the heavy and light chain variable regions, and the heavy and light chains are shown. Antibodies disclosed in the table include, MPB2D5,  CAS1.1.3, IMMU222, REA1062, and JOVI-3. MPB2D5 binds human TCRβV 20-1 (TCRβV2 per old nomenclature). CAS1.1.3 binds human TCRβV 27 (TCRβV14 per old nomenclature). IMMU 222 binds human TCRβV 6-5, TCRβV 6-6, or TCRβV 6-9 (TCRβV13.1 per old nomenclature). REA1062 binds human TCRβV 5-1). JOVI-3 binds human TCRβV 28 (TCRβV3.1 per old nomenclature). IMMU546 binds human TCRβV 2. MPB2D5 (murine), also referred to here as BJ1188, BJ1190 and REA654; or Antibody G Binds to human TCRVβ 20-1 SEQ ID NO: 1102 HC CDR1 (Kabat) SAYMH SEQ ID NO: 1103 HC CDR2 (Kabat) RIDPATGKTKYAPKFQA SEQ ID NO: 1104 HC CDR3 (Kabat) SLNWDYGLDY SEQ ID NO: 1105 HC CDR1 (Chothia) GFNIKSA SEQ ID NO: 1106 HC CDR2 (Chothia) DPATGK SEQ ID NO: 1104 HC CDR3 (Chothia) SLNWDYGLDY SEQ ID NO: 3640 HC CDR1 (Combined) GFNIKSAYMH SEQ ID NO: 1103 HC CDR2 (Combined) RIDPATGKTKYAPKFQA SEQ ID NO: 1104 HC CDR3 (Combined) SLNWDYGLDY SEQ ID NO: 1107 LC CDR1 (Kabat) RASKSVSILGTHLIH SEQ ID NO: 1108 LC CDR2 (Kabat) AASNLES SEQ ID NO: 1109 LC CDR3 (Kabat) QQSIEDPWT SEQ ID NO: 1110 LC CDR1 (Chothia) SKSVSILGTHL SEQ ID NO: 1108 LC CDR2 (Chothia) AASNLES SEQ ID NO: 1109 LC CDR3 (Chothia) QQSIEDPWT SEQ ID NO: 1107 LC CDR1 (Combined) RASKSVSILGTHLIH SEQ ID NO: 1108 LC CDR2 (Combined) AASNLES SEQ ID NO: 1109 LC CDR3 (Combined) QQSIEDPWT SEQ ID NO: 1111 VL DIVLTQSPASLAVSLGQRATISCRASKSVSILGTHLIHW YQQKPGQPPKLLIYAASNLESGVPARFSGSGSETVFTL NIHPVEEEDAATYFCQQSIEDPWTFGGGTKLGIK SEQ ID NO: 1112 VH EVQLQQSVADLVRPGASLKLSCTASGFNIKSAYMHW VIQRPDQGPECLGRIDPATGKTKYAPKFQAKATITADT SSNTAYLQLSSLTSEDTAIYYCTRSLNWDYGLDYWGQ GTSVTVSS VH for MPB2D5 (humanized) also referred to as Antibody G-H (humanized) Binds to human TCRVβ 20-1 SEQ ID NO: 1113 VH-1 QVQLVQSGAEVKKPGASVKVSCKASGFNIKSAYMHW VRQAPGQGLEWMGRIDPATGKTKYAPKFQARVTMT ADTSTNTAYMELSSLRSEDTAVYYCARSLNWDYGLD YWGQGTLVTVSS SEQ ID NO: 1114 VH-2 QVQLVQSGAEVKKPGASVKVSCKASGFNIKSAYMHW VRQAPGQEPGCMGRIDPATGKTKYAPKFQARVTMTA DTSINTAYTELSSLRSEDTATYYCARSLNWDYGLDYW GQGTLVTVSS SEQ ID NO: 1115 VH-3 QVQLVQSGAEVKKPGSSVKVSCKASGFNIKSAYMHW VRQAPGQGLEWMGRIDPATGKTKYAPKFQARVTITA DTSTNTAYMELSSLRSEDTAVYYCARSLNWDYGLDY WGQGTLVTVSS SEQ ID NO: 1116 VH-4 QVQLVQSGAEVKKPGASVKVSCKASGFNIKSAYMHW VRQAPGQRLEWMGRIDPATGKTKYAPKFQARVTITA DTSANTAYMELSSLRSEDTAVYYCARSLNWDYGLDY WGQGTLVTVSS VL for MPB2D5 (humanized) also referred to as Antibody G-H (humanized) Binds to human TCRVβ 20-1 SEQ ID NO: 1117 VL-1 EIVLTQSPATLSLSPGERATLSCRASKSVSILGTHLIHW YQQKPGQAPRLLIYAASNLESGIPARFSGSGSETDFTL TISSLEPEDFAVYFCQQSIEDPFGGGTKVEIK SEQ ID NO: 1118 VL-2 EIVLTQSPATLSLSPGERATLSCRASKSVSILGTHLIHW YQQKPGLAPRLLIYAASNLESGIPDRFSGSGSETDFTLT ISRLEPEDFAVYFCQQSIEDPFGGGTKVEIK SEQ ID NO: 1119 VL-3 EIVLTQSPGTLSLSPGERATLSCRASKSVSILGTHLIHW YQQKPGQAPRLLIYAASNLESGIPDRFSGSGSETDFTL TISRLEPEDFAVYFCQQSIEDPFGGGTKVEIK CAS1.1.3 (murine) also referred to herein as BJ1460; or Antibody H Binds to human TCRVβ 27 SEQ ID NO: 1120 HC CDR1 (Kabat) DTYMY SEQ ID NO: 1121 HC CDR2 (Kabat) RIDPANGNTKYDPKFQD SEQ ID NO: 1122 HC CDR3 (Kabat) GSYYYAMDY SEQ ID NO: 1123 HC CDR1 (Chothia) GFKTEDT SEQ ID NO: 1124 HC CDR2 (Chothia) DPANGN SEQ ID NO: 1122 HC CDR3 (Chothia) GSYYYAMDY SEQ ID NO: 1125 HC CDR1 (Combined) GFKTEDTYMY SEQ ID NO: 1121 HC CDR2 (Combined) RIDPANGNTKYDPKFQD SEQ ID NO: 1122 HC CDR3 (Combined) GSYYYAMDY SEQ ID NO: 1126 LC CDR1 (Kabat) RASESVDSYGNSFMH SEQ ID NO: 1127 LC CDR2 (Kabat) RASNLES SEQ ID NO: 1128 LC CDR3 (Kabat) QQSNEDPYT SEQ ID NO: 3641 LC CDR1 (Chothia) SESVDSYGNSF SEQ ID NO: 1127 LC CDR2 (Chothia) RASNLES SEQ ID NO: 1128 LC CDR3 (Chothia) QQSNEDPYT SEQ ID NO: 1126 LC CDR1 (Combined) RASESVDSYGNSFMH SEQ ID NO: 1127 LC CDR2 (Combined) RASNLES SEQ ID NO: 1128 LC CDR3 (Combined) QQSNEDPYT SEQ ID NO: 1129 VL DIVLTQSPASLAVSLGQRATISCRASESVDSYGNSFMH WYQQKPGQPPKLLIYRASNLESGIPARFSGSGSRTDFT LTTNPVEADDVATYYCQQSNEDPYTFGGGTKLEIK SEQ ID NO: 1130 VH EVQLQQSGAELVKPGASVKLSCTASGFKTEDTYMYW VKQRPEQGLEWIGRIDPANGNTKYDPKFQDKATITAD SSSNTAYLQLSSLPSEDTAVYYCARGSYYYAMDYWG QGTSVTVSS VH for CAS1.1.3 (humanized) also referred to as Antibody H-H (humanized) Binds to human TCRVβ 27 SEQ ID NO: 1131 VH-1 QVQLVQSGAEVKKPGSSVKVSCKASGFKTEDTYMY WVRQAPGQGLEWIGRIDPANGNTKYDPKFQDRATIT ADSSTNTAYMELSSLRSEDTAVYYCARGSYYYAMDY WGQGTLVTVSS SEQ ID NO: 1132 VH-2 QVQLVQSGAEVKKPGASVKVSCKASGFKTEDTYMY WVRQAPGQRLEWIGRIDPANGNTKYDPKFQDRATITA DSSANTAYMELSSLRSEDTAVYYCARGSYYYAMDY WGQGTLVTVSS SEQ ID NO: 1133 VH-3 EVQLVESGGGLVQPGGSLKLSCAASGFKTEDTYMYW VRQASGKGLEWIGRIDPANGNTKYDPKFQDRATISAD SSKNTAYLQMNSLKTEDTAVYYCARGSYYYAMDYW GQGTLVTVSS SEQ ID NO: 1134 VH-4 EVQLVQSGAEVKKPGESLRISCKASGFKTEDTYMYW VRQMPGKGLEWIGRIDPANGNTKYDPKFQDQATISAD SSINTAYLQWSSLKASDTAMYYCARGSYYYAMDYW GQGTLVTVSS SEQ ID NO: 1135 VH-5 QVQLVQSGSELKKPGASVKVSCKASGFKTEDTYMYW VRQAPGQGLEWIGRIDPANGNTKYDPKFQDRAVISAD SSVNTAYLQISSLKAEDTAVYYCARGSYYYAMDYWG QGTLVTVSS VL for CAS1.1.3 (humanized) also referred to as Antibody H-H (humanized) Binds to human TCRVβ 27 SEQ ID NO: 1136 VL-1 DIVLTQSPDSLAVSLGERATINCRASESVDSYGNSFMH WYQQKPGQPPKLLIYRASNLESGVPDRFSGSGSRTDF TLTISSLQAEDVAVYYCQQSNEDPYTFGQGTKLEIK SEQ ID NO: 1137 VL-2 EIVLTQSPATLSLSPGERATLSCRASESVDSYGNSFMH WYQQKPGQAPKLLIYRASNLESGIPARFSGSGSRTDFT LTISRLEPEDFAVYYCQQSNEDPYTFGQGTKLEIK SEQ ID NO: 1138 VL-3 DIQLTQSPSSLSASVGDRVTITCRASESVDSYGNSFMH WYQQKPGQAPKLLIYRASNLESGVPSRFSGSGSRTDF TLTISSLQPEDVATYYCQQSNEDPYTFGQGTKLEIK SEQ ID NO: 1139 VL-4 AIQLTQSPSSLSASVGDRVTITCRASESVDSYGNSFMH WYQQKPGKAPKLLIYRASNLESGVPSRFSGSGSRTDF TLTISSLQPEDFATYYCQQSNEDPYTFGQGTKLEIK SEQ ID NO: 1140 VL-5 EIVLTQSPDFQSVTPKEKVTITCRASESVDSYGNSFMH WYQQKPDQSPKLLIYRASNLESGVPSRFSGSGSRTDFT LTINSLEAEDAATYYCQQSNEDPYTFGQGTKLEIK IMMU222 (murine) also referred to as BJ1461; or Antibody I Binds to human TCRVβ 6-5, 6-6, 6-9 SEQ ID NO: 1141 HC CDR1 (Kabat) SYAMS SEQ ID NO: 1142 HC CDR2 (Kabat) HISNGGDYIYYADTVKG SEQ ID NO: 1143 HC CDR3 (Kabat) PSYYSDPWFFDV SEQ ID NO: 1144 HC CDR1 (Chothia) GFTFRSY SEQ ID NO: 1145 HC CDR2 (Chothia) SNGGDY SEQ ID NO: 1143 HC CDR3 (Chothia) PSYYSDPWFFDV SEQ ID NO: 1146 HC CDR1 (Combined) GFTFRSYAMS SEQ ID NO: 1142 HC CDR2 (Combined) HISNGGDYIYYADTVKG SEQ ID NO: 1143 HC CDR3 (Combined) PSYYSDPWFFDV SEQ ID NO: 1147 LC CDR1 (Kabat) SAGSSVSFMH SEQ ID NO: 1148 LC CDR2 (Kabat) DTSKLAS SEQ ID NO: 1149 LC CDR3 (Kabat) LQGSGFPLT SEQ ID NO: 1150 LC CDR1 (Chothia) GSSVSF SEQ ID NO: 1148 LC CDR2 (Chothia) DTSKLAS SEQ ID NO: 1149 LC CDR3 (Chothia) LQGSGFPLT SEQ ID NO: 1147 LC CDR1 (Combined) SAGSSVSFMH SEQ ID NO: 1148 LC CDR2 (Combined) DTSKLAS SEQ ID NO: 1149 LC CDR3 (Combined) LQGSGFPLT SEQ ID NO: 1151 VL ENVLTQSPAIMSASPGEKVTMTCSAGSSVSFMHWYQ QKSSTSPKLWIYDTSKLASGVPGRFSGSGSGNSFSLTIS SMEAEDVAIYYCLQGSGFPLTFGSGTKLEIK SEQ ID NO: 1152 VH DVKLVESGEGLVKPGGSLKLSCAASGFTFRSYAMSW VRQTPEKRLEWVAHISNGGDYIYYADTVKGRFTISRD NARNTLYLQMSSLKSEDTAMYYCTRPSYYSDPWFFD VWGTGTTVTVSS VH for IMMU222 (humanized) also referred to as Antibody I-H Binds to human TCRVβ 6-5, 6-6, 6-9 SEQ ID NO: 1153 VH-1 EVQLVESGGGLVQPGGSLRLSCAASGFTFRSYAMSW VRQAPGKGLEWVAHISNGGDYIYYADTVKGRFTISRD NAKNSLYLQMNSLRAEDTAVYYCTRPSYYSDPWFFD VWGQGTTVTVSS SEQ ID NO: 1154 VH-2 QVQLVESGGGVVQPGRSLRLSCAASGFTFRSYAMSW VRQAPGKGLEWVAHISNGGDYIYYADTVKGRFTISRD NSKNTLYLQMSSLRAEDTAVYYCTRPSYYSDPWFFD VWGQGTTVTVSS SEQ ID NO: 1155 VH-3 EVQLVESGGGLVQPGGSLRLSCAASGFTFRSYAMSW VRQAPGKGLEWVAHISNGGDYIYYADTVKGRFTISRD NSKNTLYLQMNSLRAEDTAVYYCTRPSYYSDPWFFD VWGQGTTVTVSS SEQ ID NO: 1156 VH-4 QVQLVQSGSELKKPGASVKVSCKASGFTFRSYAMSW VRQAPGQGLEWVAHISNGGDYIYYADTVKGRFVISR DNSVNTLYLQISSLKAEDTAVYYCTRPSYYSDPWFFD VWGQGTTVTVSS SEQ ID NO: 1157 VH-5 QVQLVQSGAEVKKPGASVKVSCKASGFTFRSYAMSW VRQAPGQRLEWVAHISNGGDYIYYADTVKGRFTITRD NSANTLYMELSSLRSEDTAVYYCTRPSYYSDPWFFDV WGQGTTVTVSS VL for IMMU222 (humanized)) also referred to as Antibody I-H Binds to human TCRVβ 6-5, 6-6, 6-9 SEQ ID NO: 1158 VL-1 ENVLTQSPATLSLSPGERATLSCSAGSSVSFMHWYQQ KPGQAPKLLIYDTSKLASGIPARFSGSGSGNDFTLTISS LEPEDFAVYYCLQGSGFPLTFGQGTKLEIK SEQ ID NO: 1159 VL-2 ENVLTQSPDFQSVTPKEKVTITCSAGSSVSFMHWYQQ KPDQSPKLLIYDTSKLASGVPSRFSGSGSGNDFTLTINS LEAEDAATYYCLQGSGFPLTFGQGTKLEIK SEQ ID NO: 1160 VL-3 DNQLTQSPSSLSASVGDRVTITCSAGSSVSFMHWYQQ KPGKVPKLLIYDTSKLASGVPSRFSGSGSGNDFTLTISS LQPEDVATYYCLQGSGFPLTFGQGTKLEIK SEQ ID NO: 1161 VL-4 ANQLTQSPSSLSASVGDRVTITCSAGSSVSFMHWYQQ KPGKAPKLLIYDTSKLASGVPSRFSGSGSGNDFTLTISS LQPEDFATYYCLQGSGFPLTFGQGTKLEIK SEQ ID NO: 1162 VL-5 DNVLTQSPDSLAVSLGERATINCSAGSSVSFMHWYQQ KPGQPPKLLIYDTSKLASGVPDRFSGSGSGNDFTLTISS LQAEDVAVYYCLQGSGFPLTFGQGTKLEIK REA1062 (murine), also referred to as BJ1189 or as Antibody J Binds to human TCRVβ 5-1 SEQ ID NO: 1163 HC CDR1 (Kabat) DYNIH SEQ ID NO: 1164 HC CDR2 (Kabat) YINPYNGRTGYNQKFKA SEQ ID NO: 1165 HC CDR3 (Kabat) WDGSSYFDY SEQ ID NO: 1166 HC CDR1 (Chothia) GYTFTDYNIH SEQ ID NO: 1167 HC CDR2 (Chothia) NPYNGR SEQ ID NO: 1165 HC CDR3 (Chothia) WDGSSYFDY SEQ ID NO: 1166 HC CDR1 (Combined) GYTFTDYNIH SEQ ID NO: 1164 HC CDR2 (Combined) YINPYNGRTGYNQKFKA SEQ ID NO: 1165 HC CDR3 (Combined) WDGSSYFDY SEQ ID NO: 1168 LC CDR1 (Kabat) SASSSVSYMH SEQ ID NO: 1169 LC CDR2 (Kabat) EISKLAS SEQ ID NO: 1170 LC CDR3 (Kabat) QQWNYPLLT SEQ ID NO: 1297 LC CDR1 (Chothia) SSSVSY SEQ ID NO: 1169 LC CDR2 (Chothia) EISKLAS SEQ ID NO: 1170 LC CDR3 (Chothia) QQWNYPLLT SEQ ID NO: 1168 LC CDR1 (Combined) SASSSVSYMH SEQ ID NO: 1169 LC CDR2 (Combined) EISKLAS SEQ ID NO: 1170 LC CDR3 (Combined) QQWNYPLLT SEQ ID NO: 1171 VL EIVLTQSPAITAASLGQKVTITCSASSSVSYMHWYQQK SGTSPKPWIYEISKLASGVPARFSGSGSGTSYSLTISS MEAEDAAIYYCQQWNYPLLTFGAGTKLELK SEQ ID NO: 1172 VH EVQLQQSGPVLVKPGASVRMSCKASGYTFTDYNIHW VKQSHGRSLEWVGYINPYNGRTGYNQKFKAKATLTV DKSSSTAYMDLRSLTSEDSAVYYCARWDGSSYFDYW GQGTTLTVSS VH for REA1062 (humanized) also referred to as Antibody J-H Binds to human TCRVβ 5-1 SEQ ID NO: 1173 VH-1 QVQLVQSGAEVKKPGSSVKVSCKASGYTFTDYNIHW VRQAPGQGLEWVGYINPYNGRTGYNQKFKARATLTV DKSTSTAYMELSSLRSEDTAVYYCARWDGSSYFDYW GQGTTVTVSS SEQ ID NO: 1174 VH-2 QVQLVQSGAEVKKPGASVKVSCKASGYTFTDYNIHW VRQAPGQGLEWVGYINPYNGRTGYNQKFKARATLTV DKSTSTAYMELRSLRSDDMAVYYCARWDGSSYFDY WGQGTTVTVSS SEQ ID NO: 1175 VH-3 QVQLVQSGAEVKKPGASVKVSCKASGYTFTDYNIHW VRQATGQGLEWVGYINPYNGRTGYNQKFKARATLTV NKSISTAYMELSSLRSEDTAVYYCARWDGSSYFDYW GQGTTVTVSS SEQ ID NO: 1176 VH-4 EVQLVESGGGLVQPGRSLRLSCTASGYTFTDYNIHWV RQAPGKGLEWVGYINPYNGRTGYNQKFKARATLSVD KSKSIAYLQMNSLKTEDTAVYYCARWDGSSYFDYWG QGTTVTVSS SEQ ID NO: 1177 VH-5 QVQLVQSGSELKKPGASVKVSCKASGYTFTDYNIHW VRQAPGQGLEWVGYINPYNGRTGYNQKFKARAVLSV DKSVSTAYLQISSLKAEDTAVYYCARWDGSSYFDYW GQGTTVTVSS VL for REA1062 (humanized) also referred to as Antibody J-H Binds to human TCRVβ 5-1 SEQ ID NO: 1178 VL-1 EIVLTQSPATLSLSPGERATLSCSASSSVSYMHWYQQK PGQAPKLLIYEISKLASGIPARFSGSGSGTDYTLTISSLE PEDFAVYYCQQWNYPLLTFGQGTKLEIK SEQ ID NO: 1179 VL-2 EIVLTQSPATLSLSPGERATLSCSASSSVSYMHWYQQK PGQAPKLLIYEISKLASGIPARFSGSGSGTDYTLTISRLE PEDFAVYYCQQWNYPLLTFGQGTKLEIK SEQ ID NO: 1180 VL-3 EIVLTQSPDFQSVTPKEKVTITCSASSSVSYMHWYQQK PDQSPKLLIYEISKLASGVPSRFSGSGSGTDYTLTINSLE AEDAATYYCQQWNYPLLTFGQGTKLEIK SEQ ID NO: 1181 VL-4 DIQLTQSPSFLSASVGDRVTITCSASSSVSYMHWYQQK PGKAPKLLIYEISKLASGVPSRFSGSGSGTEYTLTISSLQ PEDFATYYCQQWNYPLLTFGQGTKLEIK SEQ ID NO: 1182 VL-5 AIQLTQSPSSLSASVGDRVTITCSASSSVSYMHWYQQK PGKAPKLLIYEISKLASGVPSRFSGSGSGTDYTLTISSL QPEDFATYYCQQWNYPLLTFGQGTKLEIK SEQ ID NO: 1183 VL-6 AIRLTQSPFSLSASVGDRVTITCSASSSVSYMHWYQQK PAKAPKLFIYEISKLASGVPSRFSGSGSGTDYTLTISSL QPEDFATYYCQQWNYPLLTFGQGTKLEIK SEQ ID NO: 1184 VL-7 DIVLTQSPDSLAVSLGERATINCSASSSVSYMHWYQQ KPGQPPKLLIYEISKLASGVPDRFSGSGSGTDYTLTI SSLQAEDVAVYYCQQWNYPLLTFGQGTKLEIK JOVI-3 (murine), also referred to as BJ1187 or Antibody K Binds to human TCRVβ 28 SEQ ID NO: 1185 HC CDR1 (Kabat) GSWMN SEQ ID NO: 1186 HC CDR2 (Kabat) RIYPGDGDTDYSGKFKG SEQ ID NO: 1187 HC CDR3 (Kabat) SGYFNYVPVFDY SEQ ID NO: 1188 HC CDR1 (Chothia) GYTFSGS SEQ ID NO: 1189 HC CDR2 (Chothia) YPGDGD SEQ ID NO: 1187 HC CDR3 (Chothia) SGYFNYVPVFDY SEQ ID NO: 1190 HC CDR1 (Combined) GYTFSGSWMN SEQ ID NO: 1186 HC CDR2 (Combined) RIYPGDGDTDYSGKFKG SEQ ID NO: 1187 HC CDR3 (Combined) SGYFNYVPVFDY SEQ ID NO: 1191 LC CDR1 (Kabat) SANSTVGYIH SEQ ID NO: 1192 LC CDR2 (Kabat) TTSNLAS SEQ ID NO: 1193 LC CDR3 (Kabat) HQWSFYPT SEQ ID NO: 1194 LC CDR1 (Chothia) NSTVGY SEQ ID NO: 1192 LC CDR2 (Chothia) TTSNLAS SEQ ID NO: 1193 LC CDR3 (Chothia) HQWSFYPT SEQ ID NO: 1191 LC CDR1 (Combined) SANSTVGYIH SEQ ID NO: 1192 LC CDR2 (Combined) TTSNLAS SEQ ID NO: 1193 LC CDR3 (Combined) HQWSFYPT SEQ ID NO: 1195 VL QIVLTQSPAIMSASLGEEIALTCSANSTVGYIHWYQQK SGTSPKLLIYTTSNLASGVPSRFSGSGSGTFYSLTISS VEAEDAADYFCHQWSFYPTFGGGTKLEIK SEQ ID NO: 1196 VH QIQLQQSGPEVVKPGASVQISCKASGYTFSGSWMNW VKQRPGKGLEWIGRIYPGDGDTDYSGKFKGRATLTA DKSSSTAYMRLSSLTSEDSAVYFCARSGYFNYVPVFD YWGQGTTLSVSS VH for JOVI-3 (humanized) also referred to as Antibody K-H Binds to human TCRVβ 28 SEQ ID NO: 1197 VH-1 QIQLVQSGAEVKKPGASVKVSCKASGYTFSGSWMNW VRQAPGQGLEWIGRIYPGDGDTDYSGKFKGRATLTA DKSTSTAYMELSSLRSEDTAVYYCARSGYFNYVPVFD YWGQGTTVTVSS SEQ ID NO: 1198 VH-2 QIQLVQSGAEVKKPGSSVKVSCKASGYTFSGSWMNW VRQAPGQGLEWIGRIYPGDGDTDYSGKFKGRATLTA DKSTSTAYMELSSLRSEDTAVYYCARSGYFNYVPVFD YWGQGTTVTVSS SEQ ID NO: 1199 VH-3 EIQLVQSGAEVKKPGESLKISCKASGYTFSGSWMNWV RQMPGKGLEWIGRIYPGDGDTDYSGKFKGQATLSAD KSISTAYLQWSSLKASDTAMYYCARSGYFNYVPVFD YWGQGTTVTVSS SEQ ID NO: 1200 VH-4 QIQLVQSGSELKKPGASVKVSCKASGYTFSGSWMNW VRQAPGQGLEWIGRIYPGDGDTDYSGKFKGRAVLSA DKSVSTAYLQISSLKAEDTAVYYCARSGYFNYVPVFD YWGQGTTVTVSS SEQ ID NO: 1201 VH-5 QIQLVQSGSELKKPGASVKVSCKASGYTFSGSWMNW VRQAPGQGLEWIGRIYPGDGDTDYSGKFKGRAVLSA DKSVSMAYLQISSLKAEDTAVYYCARSGYFNYVPVF DYWGQGTTVTVSS SEQ ID NO: 1202 VH-6 EIQLVESGGGLVQPGRSLRLSCTASGYTFSGSWMNWV RQAPGKGLEWIGRIYPGDGDTDYSGKFKGRATLSAD KSKSIAYLQMNSLKTEDTAVYYCARSGYFNYVPVFD YWGQGTTVTVSS VL for JOVI-3 (humanized) also referred to as Antibody K-H Binds to human TCRVβ 28 SEQ ID NO: 1203 VL-1 EIVLTQSPATLSLSPGERATLSCSANSTVGYIHWYQQK PGQAPKLLIYTTSNLASGIPARFSGSGSGTDYTLTISSL EPEDFAVYFCHQWSFYPTFGQGTKLEIK SEQ ID NO: 1204 VL-2 DIQLTQSPSFLSASVGDRVTITCSANSTVGYIHWYQQK PGKAPKLLIYTTSNLASGVPSRFSGSGSGTEYTLTISSL QPEDFATYFCHQWSFYPTFGQGTKLEIK SEQ ID NO: 1205 VL-3 EIVLTQSPATLSLSPGERATLSCSANSTVGYIHWYQQK PGQAPKLLIYTTSNLASGIPARFSGSGPGTDYTLTISSL EPEDFAVYFCHQWSFYPTFGQGTKLEIK SEQ ID NO: 1206 VL-4 DIVLTQSPDSLAVSLGERATINCSANSTVGYIHWYQQ KPGQPPKLLIYTTSNLASGVPDRFSGSGSGTDYTLTISS LQAEDVAVYFCHQWSFYPTFGQGTKLEIK SEQ ID NO: 1207 VL-5 EIVLTQSPDFQSVTPKEKVTITCSANSTVGYIHWYQQK PDQSPKLLIYTTSNLASGVPSRFSGSGSGTDYTLTINSL EAEDAATYFCHQWSFYPTFGQGTKLEIK ZOE (murine), also referred to as BJ1538 or as Antibody L Binds to human TCRVβ 4-1, 4-2, 4-3 SEQ ID NO: 1208 HC CDR1 (Kabat) DYYMY SEQ ID NO: 1209 HC CDR2 (Kabat) TISGGGSYTYSPDSVKG SEQ ID NO: 1210 HC CDR3 (Kabat) ERDIYYGNFNAMVY SEQ ID NO: 1211 HC CDR1 (Chothia) GFTFSDY SEQ ID NO: 1212 HC CDR2 (Chothia) SGGGSY SEQ ID NO: 1210 HC CDR3 (Chothia) ERDIYYGNFNAMVY SEQ ID NO: 1213 HC CDR1 (Combined) GFTFSDYYMY SEQ ID NO: 1209 HC CDR2 (Combined) TISGGGSYTYSPDSVKG SEQ ID NO: 1210 HC CDR3 (Combined) ERDIYYGNFNAMVY SEQ ID NO: 1214 LC CDR1 (Kabat) RASKSVSTSGYSYMH SEQ ID NO: 1215 LC CDR2 (Kabat) LASNLES SEQ ID NO: 1216 LC CDR3 (Kabat) QHSRDLPWT SEQ ID NO: 1217 LC CDR1 (Chothia) SKSVSTSGYSY SEQ ID NO: 1215 LC CDR2 (Chothia) LASNLES SEQ ID NO: 1216 LC CDR3 (Chothia) QHSRDLPWT SEQ ID NO: 1214 LC CDR1 (Combined) RASKSVSTSGYSYMH SEQ ID NO: 1215 LC CDR2 (Combined) LASNLES SEQ ID NO: 1216 LC CDR3 (Combined) QHSRDLPWT SEQ ID NO: 1218 VL DIVLTQSPVSLTVSLGQRATISCRASKSVSTSGYSYMH WYQQKPGQPPKLLIYLASNLESGVPARFSGSGSGTDF TLNIHPVEEEDAATYYCQHSRDLPWTFGGGTKLEIK SEQ ID NO: 1219 VH EVQLVESGGGLVKPGGSLKLSCAASGFTFSDYYMYWV RQTPEKRLEWVATISGGGSYTYSPDSVKGRFTISRDN AKNNLYLQMSSLRSEDTAMYFCARERDIYYGNFNAM VYWGRGTSVTVSS VH for ZOE (humanized) also referred to as Antibody L-H Binds to human TCRVβ 4-1, 4-2, 4-3 SEQ ID NO: 1220 VH-1 EVQLLESGGGLVQPGGSLRLSCAASGFTFSDYYMYW VRQAPGKGLEWVATISGGGSYTYSPDSVKGRFTISRD NSKNTLYLQMNSLRAEDTAVYYCARERDIYYGNFNA MVYWGRGTLVTVSS SEQ ID NO: 1221 VH-2 EVQLVESGGGLVQPGGSLRLSCAASGFTFSDYYMYW VRQAPGKGLEWVATISGGGSYTYSPDSVKGRFTISRD NAKNSLYLQMNSLRAEDTAVYYCARERDIYYGNFNA MVYWGRGTLVTVSS SEQ ID NO: 1222 VH-3 QVQLVESGGGVVQPGRSLRLSCAASGFTFSDYYMY WVRQAPGKGLEWVATISGGGSYTYSPDSVKGRFTIS RDNSKNTLYLQMNSLRAEDTAVYYCARERDIYYGN FNAMVYWGRGTLVTVSS SEQ ID NO: 1223 VH-4 QVQLVESGGGLVKPGGSLRLSCAASGFTFSDYYMYWI RQAPGKGLEWVATISGGGSYTYSPDSVKGRFTISRDN AKNSLYLQMNSLRAEDTAVYYCARERDIYYGNFNAM VYWGRGTLVTVSS VL for ZOE (humanized) also referred to as Antibody L-H Binds to human TCRVβ 4-1, 4-2, 4-3 SEQ ID NO: 1224 VL-1 EIVLTQSPGTLSLSPGERATLSCRASKSVSTSGYSYMH WYQQKPGQAPRLLIYLASNLESGIPDRFSGSGSGTDFT LTISRLEPEDFAVYYCQHSRDLPWTFGGGTKVEIK SEQ ID NO: 1225 VL-2 EIVLTQSPATLSLSPGERATLSCRASKSVSTSGYSYMH WYQQKPGQAPRLLIYLASNLESGIPARFSGSGSGTDFT LTISSLEPEDFAVYYCQHSRDLPWTFGGGTKVEIK SEQ ID NO: 1226 VL-3 DIQLTQSPSTLSASVGDRVTITCRASKSVSTSGYSYMH WYQQKPGKAPKLLIYLASNLESGVPSRFSGSGSGTEFT LTISSLQPDDFATYYCQHSRDLPWTFGGGTKVEIK SEQ ID NO: 1227 VL-4 AIQLTQSPSSLSASVGDRVTITCRASKSVSTSGYSYMH WYQQKPGKAPKLLIYLASNLESGVPSRFSGSGSGTDF TLTISSLQPEDFATYYCQHSRDLPWTFGGGTKVEIK Anti-TCRvbl9 (murine), also referred to as BJ1465; or Antibody M Binds to human TCRVβ 19 SEQ ID NO: 1229 HC CDR1 (Kabat) GYFWN SEQ ID NO: 1230 HC CDR2 (Kabat) YISYDGSNNYNPSLKN SEQ ID NO: 1231 HC CDR3 (Kabat) PSPGTGYAVDY SEQ ID NO: 1232 HC CDR1 (Chothia) GYSITSGY SEQ ID NO: 1233 HC CDR2 (Chothia) SYDGSN SEQ ID NO: 1231 HC CDR3 (Chothia) PSPGTGYAVDY SEQ ID NO: 1234 HC CDR1 (Combined) GYSITSGYFWN SEQ ID NO: 1230 HC CDR2 (Combined) YISYDGSNNYNPSLKN SEQ ID NO: 1231 HC CDR3 (Combined) PSPGTGYAVDY SEQ ID NO: 1235 LC CDR1 (Kabat) RSSQSLVHSNGNTYLH SEQ ID NO: 1236 LC CDR2 (Kabat) KVSNRFS SEQ ID NO: 1237 LC CDR3 (Kabat) SQSTHVPFT SEQ ID NO: 1238 LC CDR1 (Chothia) SQSLVHSNGNTY SEQ ID NO: 1236 LC CDR2 (Chothia) KVSNRFS SEQ ID NO: 1237 LC CDR3 (Chothia) SQSTHVPFT SEQ ID NO: 1235 LC CDR1 (Combined) RSSQSLVHSNGNTYLH SEQ ID NO: 1236 LC CDR2 (Combined) KVSNRFS SEQ ID NO: 1237 LC CDR3 (Combined) SQSTHVPFT SEQ ID NO: 1239 VL NVVMTQTPLSLPVSLGDQASISCRSSQSLVHSNGNTYL HWYLQKPGQSPKFLIYKVSNRFSGVPDRFSGGGSGTE FTLKISRVEAEDLGVYFCSQSTHVPFTFGSGTKLEIK SEQ ID NO: 1240 VH NVQLQESGPGLVKPSQSLSLTCSVAGYSITSGYFWNW IRQFPGNKLEWMGYISYDGSNNYNPSLKNRISITRDTS KNQFFLKLNSVTTEDTATYYCASPSPGTGYAVDYWG QGTSVTVSS VH for Anti-TCRvb19 (humanized) also referred to as Antibody M-H Binds to human TCRVβ 19 SEQ ID NO: 1241 VH-1 QVQLQESGPGLVKPSETLSLTCTVSGYSITSGYFWNWI RQPPGKGLEWIGYISYDGSNNYNPSLKNRVTISRDTSK NQFSLKLSSVTAADTAVYYCASPSPGTGYAVDYWGQ GTLVTVSS SEQ ID NO: 1242 VH-2 QVQLQESGPGLVKPSETLSLTCTVSGYSITSGYFWNWI RQPPGKGLEWIGYISYDGSNNYNPSLKNRVTISRDTSK NQFSLKLSSVTAADTAVYYCASPSPGTGYAVDYWGQ GTLVTVSS SEQ ID NO: 1243 VH-3 QVQLVESGGGLVQPGGSLRLSCSVSGYSITSGYFWNW VRQAPGKGLEWVGYISYDGSNNYNPSLKNRFTISRDT SKNTFYLQMNSLRAEDTAVYYCASPSPGTGYAVDYW GQGTLVTVSS VL for Anti-TCRvb19 (humanized) also referred to as Antibody M-H Binds to human TCRVβ 19 SEQ ID NO: 1244 VL-1 VVMTQSPGTLSLSPGERATLSCRSSQSLVHSNGNTYL HWYQQKPGQAPRFLIYKVSNRFSGIPDRFSGSGSGTDF TLTISRLEPEDFAVYFCSQSTHVPFTFGQGTKLEIK SEQ ID NO: 1245 VL-2 EVVMTQSPATLSLSPGERATLSCRSSQSLVHSNGNTYL HWYQQKPGQAPRFLIYKVSNRFSGIPARFSGSGSGTDF TLTISSLEPEDFAVYFCSQSTHVPFTFGQGTKLEIK SEQ ID NO: 1246 VL-3 EVVMTQSPATLSVSPGERATLSCRSSQSLVHSNGNTY LHWYQQKPGQAPRFLIYKVSNRFSGIPARFSGSGSGTE FTLTISSLQSEDFAVYFCSQSTHVPFTFGQGTKLEIK SEQ ID NO: 1247 VL-4 DVQMTQSPSSLSASVGDRVTITCRSSQSLVHSNGNTY LHWYQQKPGKAPKFLIYKVSNRFSGVPSRFSGSGSGT DFTFTISSLQPEDIATYFCSQSTHVPFTFGQGTKLEIK BL37.2 (murine), also referred to as BJ1539 or Antibody N Binds to human TCRVβ 9 SEQ ID NO: 1248 HC CDR1 (Kabat) DYIVH SEQ ID NO: 1249 HC CDR2 (Kabat) WINTYTGTPTYADDFEG SEQ ID NO: 1250 HC CDR3 (Kabat) SWRRGIRGIGFDY SEQ ID NO: 1251 HC CDR1 (Chothia) GYTFTDY SEQ ID NO: 1252 HC CDR2 (Chothia) NTYTGT SEQ ID NO: 1250 HC CDR3 (Chothia) SWRRGIRGIGFDY SEQ ID NO: 1253 HC CDR1 (Combined) GYTFTDYIVH SEQ ID NO: 1249 HC CDR2 (Combined) WINTYTGTPTYADDFEG SEQ ID NO: 1250 HC CDR3 (Combined) SWRRGIRGIGFDY SEQ ID NO: 1254 LC CDR1 (Kabat) KASKSINKYLA SEQ ID NO: 1255 LC CDR2 (Kabat) DGSTLQS SEQ ID NO: 1256 LC CDR3 (Kabat) QQHNEYPPT SEQ ID NO: 1257 LC CDR1 (Chothia) SKSINKY SEQ ID NO: 1255 LC CDR2 (Chothia) DGSTLQS SEQ ID NO: 1256 LC CDR3 (Chothia) QQHNEYPPT SEQ ID NO: 1254 LC CDR1 (Combined) KASKSINKYLA SEQ ID NO: 1255 LC CDR2 (Combined) DGSTLQS SEQ ID NO: 1256 LC CDR3 (Combined) QQHNEYPPT SEQ ID NO: 1258 VL DVQMTQSPYNLAASPGESVSINCKASKSINKYLAWYQ QKPGKPNKLLIYDGSTLQSGIPSRFSGSGSGTDFTLT IRGLEPEDFGLYYCQQHNEYPPTFGAGTKLELK SEQ ID NO: 1259 VH QLQLVQSGPELREPGESVKISCKASGYTFTDYIVHWV KQAPGKGLKWMGWINTYTGTPTYADDFEGRFVFSLE ASASTANLQISNLKNEDTATYFCARSWRRGIRGIGFD YWGQGVMVTVSS VH for BL37.2 (humanized) also referred to as Antibody N-H Binds to human TCRVβ 9 SEQ ID NO: 1260 VH-1 QLQLVQSGAEVKKPGASVKVSCKASGYTFTDYIVHW VRQAPGQGLEWMGWINTYTGTPTYADDFEGWVTMTL DASISTAYMELSRLRSDDTAVYYCARSWRRGIRGIG FDYWGQGTMVTVSS SEQ ID NO: 1261 VH-2 QLQLVQSGAEVKKPGASVKVSCKASGYTFTDYIVHW VRQAPGQGLEWMGWINTYTGTPTYADDFEGRVTMTL DASTSTAYMELSSLRSEDTAVYYCARSWRRGIRGIG FDYWGQGTMVTVSS SEQ ID NO: 1262 VH-3 QLQLVQSGAEVKKPGASVKVSCKASGYTFTDYIVHW VRQAPGQRLEWMGWINTYTGTPTYADDFEGRVTITL DASASTAYMELSSLRSEDMAVYYCARSWRRGIRGIG FDYWGQGTMVTVSS SEQ ID NO: 1263 VH-4 QLQLVQSGAEVKKPGASVKVSCKASGYTFTDYIVHW VRQATGQGLEWMGWINTYTGTPTYADDFEGRVTMTL NASISTAYMELSSLRSEDTAVYYCARSWRRGIRGIG FDYWGQGTMVTVSS VL for BL37.2 (humanized) also referred to as Antibody N-H Binds to human TCRVβ 9 SEQ ID NO: 1264 VL-1 EVVMTQSPGTLSLSPGERATLSCKASKSINKYLAWYQ QKPGQAPRLLIYDGSTLQSGIPDRFSGSGSGTDFTLTIS RLEPEDFAVYYCQQHNEYPPTFGQGTKLEIK SEQ ID NO: 1265 VL-2 EVVMTQSPATLSLSPGERATLSCKASKSINKYLAWYQ QKPGQAPRLLIYDGSTLQSGIPARFSGSGSGTDFTLTIS SLEPEDFAVYYCQQHNEYPPTFGQGTKLEIK SEQ ID NO: 1266 VL-3 DVQMTQSPSSLSASVGDRVTITCKASKSINKYLAWYQ QKPGKAPKLLIYDGSTLQSGVPSRFSGSGSGTDFTLTIS SLQPEDFATYYCQQHNEYPPTFGQGTKLEIK SEQ ID NO: 1267 VL-4 AVRMTQSPSSFSASTGDRVTITCKASKSINKYLAWYQ QKPGKAPKLLIYDGSTLQSGVPSRFSGSGSGTDFTLTIS CLQSEDFATYYCQQHNEYPPTFGQGTKLEIK IG125 (murine) binds to TRVβ 11-2; also referred to as Antibody O SEQ ID NO: 1268 HC CDR1 (Kabat) NYGVH SEQ ID NO: 1269 HC CDR2 (Kabat) VIWSDGSTDYDTAFIS SEQ ID NO: 1270 HC CDR3 (Kabat) RAVVADFDY SEQ ID NO: 1271 HC CDR1 (Chothia) GFSLTN SEQ ID NO: 1272 HC CDR2 (Chothia) VIWSDGSTD SEQ ID NO: 1270 HC CDR3 (Chothia) RAVVADFDY SEQ ID NO: 1273 HC CDR1 (combined) GFSLTNYGVH SEQ ID NO: 1269 HC CDR2 (combined) VIWSDGSTDYDTAFIS SEQ ID NO: 1270 HC CDR3 (combined) RAVVADFDY SEQ ID NO: 1274 VH QVQLKQSGPGLLQPSQSLSITCTVSGFSLTNYGVHWV RQSPGKGLEWLGVIWSDGSTDYDTAFISRLSISKDNSK SQVFFKLNSLQADDTAIYYCARRAVVADFDYWGQGT TLTVSS SEQ ID NO: 1275 LC CDR1 (Kabat) KASKEVTIFGSISALH SEQ ID NO: 1276 LC CDR2 (Kabat) NGAKLES SEQ ID NO: 1277 LC CDR3 (Kabat) LQNKEVPFT SEQ ID NO: 1275 LC CDR1 (Chothia) KASKEVTIFGSISALH SEQ ID NO: 1276 LC CDR2 (Chothia) NGAKLES SEQ ID NO: 1277 LC CDR3 (Chothia) LQNKEVPFT SEQ ID NO: 1275 LC CDR1 (combined) KASKEVTIFGSISALH SEQ ID NO: 1276 LC CDR2 (combined) NGAKLES SEQ ID NO: 1277 LC CDR3 (combined) LQNKEVPFT SEQ ID NO: 1278 VL DIVLTQSPASLAVSLGQKATISCKASKEVTIFGSISA LHWYQQKPGQPPKLIYNGAKLESGVSARFSDSGSQNR SPFGNQLSFTLTIAPVEADDAATYYCLQNKEVPFTFG SGTKLEIK VL for IG125 (humanized) also referred to as Antibody O-H binds to TRVβ 11-2 SEQ ID NO: 1279 VL-1 DIVLTQSPDSLAVSLGERATINCKASKEVTIFGSISALH WYQQKPGQPPKLLYNGAKLESGVSARFGVPDRFSRS GSGLDFTLTISSLQAEDVAVYYCLQNKEVPFTFGQGT KLEIK SEQ ID NO: 1280 VL-2 EIVLTQSPDFQSVTPKEKVTITCKASKEVTIFGSISALH WYQQKPDQSPKLLYNGAKLESGVSARFGVPSRFSRS GSGLDFTLTINSLEAEDAATYYCLQNKEVPFTFGQGT KLEIK SEQ ID NO: 1281 VL-3 AIQLTQSPSSLSASVGDRVTITCKASKEVTIFGSISALH WYQQKPGKAPKLLYNGAKLESGVSARFGVPSRFSRS GSGLDFTLTISSLQPEDFATYYCLQNKEVPFTFGQGT KLEIK SEQ ID NO: 1282 VL-4 DIVLTQTPLSLSVTPGQPASISCKASKEVTIFGSISALH WYLQKPGQPPKLLYNGAKLESGVSARFGVPDRFSRS GSGLDFTLKISRVEAEDVGVYYCLQNKEVPFTFGQG TKLEIK VH for IG125 (humanized) also referred to as Antibody O-H binds to TRVβ 11-2 SEQ ID NO: 1283 VH-1 QVTLKESGPVLVKPTETLTLTCTVSGFSLTNYGVHW VRQPPGKALEWLGVIWSDGSTDYDTAFISRLTISKDN SKSQVVLTMTNMDPVDTATYYCARRAVVADFDYW GQGTTVTVSS SEQ ID NO: 1284 VH-2 QVQLQESGPGLVKPSGTLSLTCAVSGFSLTNYGVHW VRQPPGKGLEWLGVIWSDGSTDYDTAFISRLTISKDN SKSQVSLKLSSVTAADTAVYYCARRAVVADFDYWG QGTTVTVSS SEQ ID NO: 1285 VH-3 QVQLQQSGPGLVKPSQTLSLTCAVSGFSLTNYGVHW VRQSPSRGLEWLGVIWSDGSTDYDTAFISRLTINKDN SKSQVSLQLNSVTPEDTAVYYCARRAVVADFDYWG QGTTVTVSS SEQ ID NO: 1286 VH-4 EVQLVESGGGLVQPGPSLRLSCTVSGFSLTNYGVHW VRQAPGKGLEWLGVIWSDGSTDYDTAFISRLTISKDN SKSIVYLQMNSLKTEDTAVYYCARRAVVADFDYWG QGTTVTVSS SEQ ID NO: 1287 VH-5 EVQLVQSGAEVKKPGESLRISCKVSGFSLTNYGVHW VRQMPGKGLEWLGVIWSDGSTDYDTAFISQLTISKD NSISTVYLQWSSLKASDTAMYYCARRAVVADFDYW GQGTTVTVSS MR5-2 (murine), binds to human TCRVβ 13-2 SEQ ID NO: 1376 SCFV (VH + VL) QVQLQQSGTELMKPGASVKISCKASGYTFSNYWIEW IKQRPGHGLEWVGEILPGAGPTNYNEKFKGKATFTA DSSSNTAYMQLSSLTSEDSAVYYCARTDYDYDWFA YWGQGTLVTVSAGGGGSGGGGSGGGGSGGGGSDIV MSQSPSSLAVSVGEKVTMSCKSSQSLLYSGNQKNYL AWYQQKPGQSPKLLIYWASTRESGVPDRFTGSGSGT DFTLTINSVKAEDLTVYYCQQYYGYPRTFGGGTKVEI K

Anti-TCRVβ Antibody Effector Function and Fc Variants

In some embodiments, an anti-TCRVβ antibody disclosed herein comprises an Fc region, e.g., as described herein. In some embodiments, the Fc region is a wildtype Fc region, e.g., a wildtype human Fc region. In some embodiments, the Fc region comprises a variant, e.g., an Fc region comprising an addition, substitution, or deletion of at least one amino acid residue in the Fc region which results in, e.g., reduced or ablated affinity for at least one Fc receptor.

The Fc region of an antibody interacts with a number of receptors or ligands including Fc Receptors (e.g., FcγRT, FcγRIIA, FcγRIIIA), the complement protein CIq, and other molecules such as proteins A and G. These interactions are essential for a variety of effector functions and downstream signaling events including: antibody dependent cell-mediated cytotoxicity (ADCC), Antibody-dependent cellular phagocytosis (ADCP) and complement dependent cytotoxicity (CDC).

In some embodiments, an anti-TCRVβ antibody comprising a variant Fc region has reduced, e.g., ablated, affinity for an Fc receptor, e.g., an Fc receptor described herein. In some embodiments, the reduced affinity is compared to an otherwise similar antibody with a wildtype Fc region.

In some embodiments, an anti-TCRVβ antibody comprising a variant Fc region has one or more of the following properties: (1) reduced effector function (e.g., reduced ADCC, ADCP and/or CDC); (2) reduced binding to one or more Fc receptors; and/or (3) reduced binding to C1q complement. In some embodiments, the reduction in any one, or all of properties (1)-(3) is compared to an otherwise similar antibody with a wildtype Fc region.

In some embodiments, an anti-TCRVβ antibody comprising a variant Fc region has reduced affinity to a human Fc receptor, e.g., FcγR I, FcγR II and/or FcγR III. In some embodiments, the anti-TCRVβ antibody comprising a variant Fc region comprises a human IgG1 region or a human IgG4 region.

In some embodiments, an anti-TCRVβ antibody comprising a variant Fc region activates and/or expands T cells, e.g., as described herein. In some embodiments, an anti-TCRVβ antibody comprising a variant Fc region has a cytokine profile described herein, e.g., a cytokine profile that differs from a cytokine profile of a T cell engager that binds to a receptor or molecule other than a TCRβV region (“a non-TCRβV-binding T cell engager”). In some embodiments, the non-TCRβV-binding T cell engager comprises an antibody that binds to a CD3 molecule (e.g., CD3 epsilon (CD3e) molecule); or a TCR alpha (TCRα) molecule.

Exemplary Fc region variants are provided in Table 21 and also disclosed in Saunders 0, (2019) Frontiers in Immunology; vol 10, article 1296, the entire contents of which is hereby incorporated by reference.

In some embodiments, an anti-TCRVβ antibody disclosed herein comprises any one or all, or any combination of Fc region variants disclosed in Table 21.

In some embodiments, an anti-TCRVβ antibody disclosed herein comprises any one or all, or any combination of Fc region variants, e.g., mutations, disclosed in Table 21. In some embodiments, an anti-TCRVβ antibody disclosed herein comprise an Asn297Ala (N297A) mutation. In some embodiments, an anti-TCRVβ antibody disclosed herein comprise a Leu234A1a/Leu235Ala (LALA) mutation.

TABLE 21 Exemplary Fc modifications Altered effector Modification or mutation function Leu235Glu ADCC; Leu234Ala/Leu235Ala (LALA) ADCC; ADCP; CDC Ser228Pro/Leu235Glu Leu234Ala/Leu235Ala/Pro329Gly ADCP Pro331Ser/Leu234Glu/Leu235Phe CDC Asp265Ala ADCC, ADCP Gly237Ala ADCP Glu318Ala ADCP Glu233Pro Gly236Arg/Leu328Arg ADCC His268Gln/Val309Leu/Ala330Ser/Pro331Ser ADCC; ADCP; CDC Val234Ala/Gly237Ala/Pro238Ser/ ADCC; ADCP; CDC His268Ala/Val309Leu/Ala330Ser/Pro331Ser Leu234Ala/L235Ala/Gly237Ala/P238Ser/ ADCC; CDC His268Ala/Ala330Ser/Pro331Ser Ala330Leu CDC Asp270Ala CDC Lys322Ala CDC Pro329Ala CDC Pro331Ala CDC Val264Ala CDC High mannose glycosylation CDC Phe241Ala CDC Asn297Ala or Gly or Gln ADCC; ADCP; CDC S228P/Phe234Ala/Leu235Ala ADCC; CDC

Antibody Molecules

In one embodiment, the antibody molecule binds to a cancer antigen, e.g., a tumor antigen or a stromal antigen. In some embodiments, the cancer antigen is, e.g., a mammalian, e.g., a human, cancer antigen. In other embodiments, the antibody molecule binds to an immune cell antigen, e.g., a mammalian, e.g., a human, immune cell antigen. For example, the antibody molecule binds specifically to an epitope, e.g., linear or conformational epitope, on the cancer antigen or the immune cell antigen.

In an embodiment, an antibody molecule is a monospecific antibody molecule and binds a single epitope. E.g., a monospecific antibody molecule having a plurality of immunoglobulin variable domain sequences, each of which binds the same epitope.

In an embodiment an antibody molecule is a multispecific or multifunctional antibody molecule, e.g., it comprises a plurality of immunoglobulin variable domains sequences, wherein a first immunoglobulin variable domain sequence of the plurality has binding specificity for a first epitope and a second immunoglobulin variable domain sequence of the plurality has binding specificity for a second epitope. In an embodiment the first and second epitopes are on the same antigen, e.g., the same protein (or subunit of a multimeric protein). In an embodiment the first and second epitopes overlap. In an embodiment the first and second epitopes do not overlap. In an embodiment the first and second epitopes are on different antigens, e.g., the different proteins (or different subunits of a multimeric protein). In an embodiment a multispecific antibody molecule comprises a third, fourth or fifth immunoglobulin variable domain. In an embodiment, a multispecific antibody molecule is a bispecific antibody molecule, a trispecific antibody molecule, or a tetraspecific antibody molecule.

In an embodiment a multispecific antibody molecule is a bispecific antibody molecule. A bispecific antibody has specificity for no more than two antigens. A bispecific antibody molecule is characterized by a first immunoglobulin variable domain sequence which has binding specificity for a first epitope and a second immunoglobulin variable domain sequence that has binding specificity for a second epitope. In an embodiment the first and second epitopes are on the same antigen, e.g., the same protein (or subunit of a multimeric protein). In an embodiment the first and second epitopes overlap. In an embodiment the first and second epitopes do not overlap. In an embodiment the first and second epitopes are on different antigens, e.g., the different proteins (or different subunits of a multimeric protein). In an embodiment a bispecific antibody molecule comprises a heavy chain variable domain sequence and a light chain variable domain sequence which have binding specificity for a first epitope and a heavy chain variable domain sequence and a light chain variable domain sequence which have binding specificity for a second epitope. In an embodiment a bispecific antibody molecule comprises a half antibody having binding specificity for a first epitope and a half antibody having binding specificity for a second epitope. In an embodiment a bispecific antibody molecule comprises a half antibody, or fragment thereof, having binding specificity for a first epitope and a half antibody, or fragment thereof, having binding specificity for a second epitope. In an embodiment a bispecific antibody molecule comprises a scFv or a Fab, or fragment thereof, have binding specificity for a first epitope and a scFv or a Fab, or fragment thereof, have binding specificity for a second epitope.

In an embodiment, an antibody molecule comprises a diabody, and a single-chain molecule, as well as an antigen-binding fragment of an antibody (e.g., Fab, F(ab′)2, and Fv). For example, an antibody molecule can include a heavy (H) chain variable domain sequence (abbreviated herein as VH), and a light (L) chain variable domain sequence (abbreviated herein as VL). In an embodiment an antibody molecule comprises or consists of a heavy chain and a light chain (referred to herein as a half antibody. In another example, an antibody molecule includes two heavy (H) chain variable domain sequences and two light (L) chain variable domain sequence, thereby forming two antigen binding sites, such as Fab, Fab′, F(ab′)2, Fc, Fd, Fd′, Fv, single chain antibodies (scFv for example), single variable domain antibodies, diabodies (Dab) (bivalent and bispecific), and chimeric (e.g., humanized) antibodies, which may be produced by the modification of whole antibodies or those synthesized de novo using recombinant DNA technologies. These functional antibody fragments retain the ability to selectively bind with their respective antigen or receptor. Antibodies and antibody fragments can be from any class of antibodies including, but not limited to, IgG, IgA, IgM, IgD, and IgE, and from any subclass (e.g., IgG1, IgG2, IgG3, and IgG4) of antibodies. The preparation of antibody molecules can be monoclonal or polyclonal. An antibody molecule can also be a human, humanized, CDR-grafted, or in vitro generated antibody. The antibody can have a heavy chain constant region chosen from, e.g., IgG1, IgG2, IgG3, or IgG4. The antibody can also have a light chain chosen from, e.g., kappa or lambda. The term “immunoglobulin” (Ig) is used interchangeably with the term “antibody” herein.

Examples of antigen-binding fragments of an antibody molecule include: (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab′)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a diabody (dAb) fragment, which consists of a VH domain; (vi) a camelid or camelized variable domain; (vii) a single chain Fv (scFv), see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883); (viii) a single domain antibody. These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies.

Antibody molecules include intact molecules as well as functional fragments thereof. Constant regions of the antibody molecules can be altered, e.g., mutated, to modify the properties of the antibody (e.g., to increase or decrease one or more of: Fc receptor binding, antibody glycosylation, the number of cysteine residues, effector cell function, or complement function).

Antibody molecules can also be single domain antibodies. Single domain antibodies can include antibodies whose complementary determining regions are part of a single domain polypeptide. Examples include, but are not limited to, heavy chain antibodies, antibodies naturally devoid of light chains, single domain antibodies derived from conventional 4-chain antibodies, engineered antibodies and single domain scaffolds other than those derived from antibodies. Single domain antibodies may be any of the art, or any future single domain antibodies. Single domain antibodies may be derived from any species including, but not limited to mouse, human, camel, llama, fish, shark, goat, rabbit, and bovine. According to another aspect of the invention, a single domain antibody is a naturally occurring single domain antibody known as heavy chain antibody devoid of light chains. Such single domain antibodies are disclosed in WO 9404678, for example. For clarity reasons, this variable domain derived from a heavy chain antibody naturally devoid of light chain is known herein as a VHH or nanobody to distinguish it from the conventional VH of four chain immunoglobulins. Such a VHH molecule can be derived from antibodies raised in Camelidae species, for example in camel, llama, dromedary, alpaca and guanaco. Other species besides Camelidae may produce heavy chain antibodies naturally devoid of light chain; such VHHs are within the scope of the invention.

The VH and VL regions can be subdivided into regions of hypervariability, termed “complementarity determining regions” (CDR), interspersed with regions that are more conserved, termed “framework regions” (FR or FW).

The extent of the framework region and CDRs has been precisely defined by a number of methods (see, Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242; Chothia, C. et al. (1987) J. Mol. Biol. 196:901-917; and the AbM definition used by Oxford Molecular's AbM antibody modeling software. See, generally, e.g., Protein Sequence and Structure Analysis of Antibody Variable Domains. In: Antibody Engineering Lab Manual (Ed.: Duebel, S. and Kontermann, R., Springer-Verlag, Heidelberg).

The terms “complementarity determining region,” and “CDR,” as used herein refer to the sequences of amino acids within antibody variable regions which confer antigen specificity and binding affinity. In general, there are three CDRs in each heavy chain variable region (HCDR1, HCDR2, HCDR3) and three CDRs in each light chain variable region (LCDR1, LCDR2, LCDR3).

The precise amino acid sequence boundaries of a given CDR can be determined using any of a number of known schemes, including those described by Kabat et al. (1991), “Sequences of Proteins of Immunological Interest,” 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD (“Kabat” numbering scheme), Al-Lazikani et al., (1997) JMB 273,927-948 (“Chothia” numbering scheme). As used herein, the CDRs defined according the “Chothia” number scheme are also sometimes referred to as “hypervariable loops.”

For example, under Kabat, the CDR amino acid residues in the heavy chain variable domain (VH) are numbered 31-35 (HCDR1), 50-65 (HCDR2), and 95-102 (HCDR3); and the CDR amino acid residues in the light chain variable domain (VL) are numbered 24-34 (LCDR1), 50-56 (LCDR2), and 89-97 (LCDR3). Under Chothia, the CDR amino acids in the VH are numbered 26-32 (HCDR1), 52-56 (HCDR2), and 95-102 (HCDR3); and the amino acid residues in VL are numbered 26-32 (LCDR1), 50-52 (LCDR2), and 91-96 (LCDR3).

Each VH and VL typically includes three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.

The antibody molecule can be a polyclonal or a monoclonal antibody.

The terms “monoclonal antibody” or “monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of single molecular composition. A monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope. A monoclonal antibody can be made by hybridoma technology or by methods that do not use hybridoma technology (e.g., recombinant methods).

The antibody can be recombinantly produced, e.g., produced by phage display or by combinatorial methods, or by yeast display.

Phage display and combinatorial methods for generating antibodies are known in the art (as described in, e.g., Ladner et al. U.S. Pat. No. 5,223,409; Kang et al. International Publication No. WO 92/18619; Dower et al. International Publication No. WO 91/17271; Winter et al. International Publication WO 92/20791; Markland et al. International Publication No. WO 92/15679; Breitling et al. International Publication WO 93/01288; McCafferty et al. International Publication No. WO 92/01047; Garrard et al. International Publication No. WO 92/09690; Ladner et al. International Publication No. WO 90/02809; Fuchs et al. (1991) Bio/Technology 9:1370-1372; Hay et al. (1992) Hum Antibod Hybridomas 3:81-85; Huse et al. (1989) Science 246:1275-1281; Griffths et al. (1993) EMBO J 12:725-734; Hawkins et al. (1992) J Mol Biol 226:889-896; Clackson et al. (1991) Nature 352:624-628; Gram et al. (1992) PNAS 89:3576-3580; Garrad et al. (1991) Bio/Technology 9:1373-1377; Hoogenboom et al. (1991) Nuc Acid Res 19:4133-4137; and Barbas et al. (1991) PNAS 88:7978-7982, the contents of all of which are incorporated by reference herein).

The yeast display method for generating or identifying antibodies is known in the art, e.g., as described in Chao et al. (2006) Nature Protocols 1(2):755-68, the entire contents of which is incorporated by reference herein.

In one embodiment, the antibody is a fully human antibody (e.g., an antibody made in a mouse which has been genetically engineered to produce an antibody from a human immunoglobulin sequence), or a non-human antibody, e.g., a rodent (mouse or rat), goat, primate (e.g., monkey), camel antibody. Preferably, the non-human antibody is a rodent (mouse or rat antibody). Methods of producing rodent antibodies are known in the art.

Human monoclonal antibodies can be generated using transgenic mice carrying the human immunoglobulin genes rather than the mouse system. Splenocytes from these transgenic mice immunized with the antigen of interest are used to produce hybridomas that secrete human mAbs with specific affinities for epitopes from a human protein (see, e.g., Wood et al. International Application WO 91/00906, Kucherlapati et al. PCT publication WO 91/10741; Lonberg et al. International Application WO 92/03918; Kay et al. International Application 92/03917; Lonberg, N. et al. 1994 Nature 368:856-859; Green, L. L. et al. 1994 Nature Genet. 7:13-21; Morrison, S. L. et al. 1994 Proc. Natl. Acad. Sci. USA 81:6851-6855; Bruggeman et al. 1993 Year Immunol 7:33-40; Tuaillon et al. 1993 PNAS 90:3720-3724; Bruggeman et al. 1991 Eur J Immunol 21:1323-1326).

An antibody molecule can be one in which the variable region, or a portion thereof, e.g., the CDRs, are generated in a non-human organism, e.g., a rat or mouse. Chimeric, CDR-grafted, and humanized antibodies are within the invention. Antibody molecules generated in a non-human organism, e.g., a rat or mouse, and then modified, e.g., in the variable framework or constant region, to decrease antigenicity in a human are within the invention.

An “effectively human” protein is a protein that does substantially not evoke a neutralizing antibody response, e.g., the human anti-murine antibody (HAMA) response. HAMA can be problematic in a number of circumstances, e.g., if the antibody molecule is administered repeatedly, e.g., in treatment of a chronic or recurrent disease condition. A HAMA response can make repeated antibody administration potentially ineffective because of an increased antibody clearance from the serum (see, e.g., Saleh et al., Cancer Immunol. Immunother., 32:180-190 (1990)) and also because of potential allergic reactions (see, e.g., LoBuglio et al., Hybridoma, 5:5117-5123 (1986)).

Chimeric antibodies can be produced by recombinant DNA techniques known in the art (see Robinson et al., International Patent Publication PCT/US86/02269; Akira, et al., European Patent Application 184,187; Taniguchi, M., European Patent Application 171,496; Morrison et al., European Patent Application 173,494; Neuberger et al., International Application WO 86/01533; Cabilly et al. U.S. Pat. No. 4,816,567; Cabilly et al., European Patent Application 125,023; Better et al. (1988 Science 240:1041-1043); Liu et al. (1987) PNAS 84:3439-3443; Liu et al., 1987, J. Immunol. 139:3521-3526; Sun et al. (1987) PNAS 84:214-218; Nishimura et al., 1987, Canc. Res. 47:999-1005; Wood et al. (1985) Nature 314:446-449; and Shaw et al., 1988, J. Natl Cancer Inst. 80:1553-1559).

A humanized or CDR-grafted antibody will have at least one or two but generally all three recipient CDRs (of heavy and or light immuoglobulin chains) replaced with a donor CDR. The antibody may be replaced with at least a portion of a non-human CDR or only some of the CDRs may be replaced with non-human CDRs. It is only necessary to replace the number of CDRs required for binding to the antigen. Preferably, the donor will be a rodent antibody, e.g., a rat or mouse antibody, and the recipient will be a human framework or a human consensus framework. Typically, the immunoglobulin providing the CDRs is called the “donor” and the immunoglobulin providing the framework is called the “acceptor.” In one embodiment, the donor immunoglobulin is a non-human (e.g., rodent). The acceptor framework is a naturally-occurring (e.g., a human) framework or a consensus framework, or a sequence about 85% or higher, preferably 90%, 95%, 99% or higher identical thereto.

As used herein, the term “consensus sequence” refers to the sequence formed from the most frequently occurring amino acids (or nucleotides) in a family of related sequences (See e.g., Winnaker, From Genes to Clones (Verlagsgesellschaft, Weinheim, Germany 1987). In a family of proteins, each position in the consensus sequence is occupied by the amino acid occurring most frequently at that position in the family. If two amino acids occur equally frequently, either can be included in the consensus sequence. A “consensus framework” refers to the framework region in the consensus immunoglobulin sequence.

An antibody molecule can be humanized by methods known in the art (see e.g., Morrison, S. L., 1985, Science 229:1202-1207, by Oi et al., 1986, BioTechniques 4:214, and by Queen et al. U.S. Pat. Nos. 5,585,089, 5,693,761 and 5,693,762, the contents of all of which are hereby incorporated by reference).

Humanized or CDR-grafted antibody molecules can be produced by CDR-grafting or CDR substitution, wherein one, two, or all CDRs of an immunoglobulin chain can be replaced. See e.g., U.S. Pat. No. 5,225,539; Jones et al. 1986 Nature 321:552-525; Verhoeyan et al. 1988 Science 239:1534; Beidler et al. 1988 J. Immunol. 141:4053-4060; Winter U.S. Pat. No. 5,225,539, the contents of all of which are hereby expressly incorporated by reference. Winter describes a CDR-grafting method which may be used to prepare the humanized antibodies of the present invention (UK Patent Application GB 2188638A, filed on Mar. 26, 1987; Winter U.S. Pat. No. 5,225,539), the contents of which is expressly incorporated by reference.

Also within the scope of the invention are humanized antibody molecules in which specific amino acids have been substituted, deleted or added. Criteria for selecting amino acids from the donor are described in U.S. Pat. No. 5,585,089, e.g., columns 12-16 of U.S. Pat. No. 5,585,089, e.g., columns 12-16 of U.S. Pat. No. 5,585,089, the contents of which are hereby incorporated by reference. Other techniques for humanizing antibodies are described in Padlan et al. EP 519596 A1, published on Dec. 23, 1992.

The antibody molecule can be a single chain antibody. A single-chain antibody (scFV) may be engineered (see, for example, Colcher, D. et al. (1999) Ann N Y Acad Sci 880:263-80; and Reiter, Y. (1996) Clin Cancer Res 2:245-52). The single chain antibody can be dimerized or multimerized to generate multivalent antibodies having specificities for different epitopes of the same target protein.

In yet other embodiments, the antibody molecule has a heavy chain constant region chosen from, e.g., the heavy chain constant regions of IgG1, IgG2, IgG3, IgG4, IgM, IgA1, IgA2, IgD, and IgE; particularly, chosen from, e.g., the (e.g., human) heavy chain constant regions of IgG1, IgG2, IgG3, and IgG4. In another embodiment, the antibody molecule has a light chain constant region chosen from, e.g., the (e.g., human) light chain constant regions of kappa or lambda. The constant region can be altered, e.g., mutated, to modify the properties of the antibody (e.g., to increase or decrease one or more of: Fc receptor binding, antibody glycosylation, the number of cysteine residues, effector cell function, and/or complement function). In one embodiment the antibody has: effector function; and can fix complement. In other embodiments the antibody does not; recruit effector cells; or fix complement. In another embodiment, the antibody has reduced or no ability to bind an Fc receptor. For example, it is a isotype or subtype, fragment or other mutant, which does not support binding to an Fc receptor, e.g., it has a mutagenized or deleted Fc receptor binding region.

Methods for altering an antibody constant region are known in the art. Antibodies with altered function, e.g. altered affinity for an effector ligand, such as FcR on a cell, or the Cl component of complement can be produced by replacing at least one amino acid residue in the constant portion of the antibody with a different residue (see e.g., EP 388,151 A1, U.S. Pat. Nos. 5,624,821 and 5,648,260, the contents of all of which are hereby incorporated by reference). Similar type of alterations could be described which if applied to the murine, or other species immunoglobulin would reduce or eliminate these functions.

An antibody molecule can be derivatized or linked to another functional molecule (e.g., another peptide or protein). As used herein, a “derivatized” antibody molecule is one that has been modified. Methods of derivatization include but are not limited to the addition of a fluorescent moiety, a radionucleotide, a toxin, an enzyme or an affinity ligand such as biotin. Accordingly, the antibody molecules of the invention are intended to include derivatized and otherwise modified forms of the antibodies described herein, including immunoadhesion molecules. For example, an antibody molecule can be functionally linked (by chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more other molecular entities, such as another antibody (e.g., a bispecific antibody or a diabody), a detectable agent, a cytotoxic agent, a pharmaceutical agent, and/or a protein or peptide that can mediate association of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag).

One type of derivatized antibody molecule is produced by crosslinking two or more antibodies (of the same type or of different types, e.g., to create bispecific antibodies). Suitable crosslinkers include those that are heterobifunctional, having two distinctly reactive groups separated by an appropriate spacer (e.g., m-maleimidobenzoyl-N-hydroxysuccinimide ester) or homobifunctional (e.g., disuccinimidyl suberate). Such linkers are available from Pierce Chemical Company, Rockford, Ill.

Multispecific or Multifunctional Antibody Molecules

Exemplary structures of multispecific and multifunctional molecules defined herein are described throughout. Exemplary structures are further described in: Weidle U et al. (2013) The Intriguing Options of Multispecific Antibody Formats for Treatment of Cancer. Cancer Genomics & Proteomics 10: 1-18 (2013); and Spiess C et al. (2015) Alternative molecular formats and therapeutic applications for bispecific antibodies. Molecular Immunology 67: 95-106; the full contents of each of which is incorporated by reference herein).

In embodiments, multispecific antibody molecules can comprise more than one antigen-binding site, where different sites are specific for different antigens. In embodiments, multispecific antibody molecules can bind more than one (e.g., two or more) epitopes on the same antigen. In embodiments, multispecific antibody molecules comprise an antigen-binding site specific for a target cell (e.g., cancer cell) and a different antigen-binding site specific for an immune effector cell. In one embodiment, the multispecific antibody molecule is a bispecific antibody molecule. Bispecific antibody molecules can be classified into five different structural groups: (i) bispecific immunoglobulin G (BsIgG); (ii) IgG appended with an additional antigen-binding moiety; (iii) bispecific antibody fragments; (iv) bispecific fusion proteins; and (v) bispecific antibody conjugates.

BsIgG is a format that is monovalent for each antigen. Exemplary BsIgG formats include but are not limited to crossMab, DAF (two-in-one), DAF (four-in-one), DutaMab, DT-IgG, knobs-in-holes common LC, knobs-in-holes assembly, charge pair, Fab-arm exchange, SEEDbody, triomab, LUZ-Y, Fcab, κλ-body, orthogonal Fab. See Spiess et al. Mol. Immunol. 67(2015):95-106. Exemplary BsIgGs include catumaxomab (Fresenius Biotech, Trion Pharma, Neopharm), which contains an anti-CD3 arm and an anti-EpCAM arm; and ertumaxomab (Neovii Biotech, Fresenius Biotech), which targets CD3 and HER2. In some embodiments, BsIgG comprises heavy chains that are engineered for heterodimerization. For example, heavy chains can be engineered for heterodimerization using a “knobs-into-holes” strategy, a SEED platform, a common heavy chain (e.g., in κλ-bodies), and use of heterodimeric Fc regions. See Spiess et al. Mol. Immunol. 67(2015):95-106. Strategies that have been used to avoid heavy chain pairing of homodimers in BsIgG include knobs-in-holes, duobody, azymetric, charge pair, HA-TF, SEEDbody, and differential protein A affinity. See Id. BsIgG can be produced by separate expression of the component antibodies in different host cells and subsequent purification/assembly into a BsIgG. BsIgG can also be produced by expression of the component antibodies in a single host cell. BsIgG can be purified using affinity chromatography, e.g., using protein A and sequential pH elution.

IgG appended with an additional antigen-binding moiety is another format of bispecific antibody molecules. For example, monospecific IgG can be engineered to have bispecificity by appending an additional antigen-binding unit onto the monospecific IgG, e.g., at the N- or C-terminus of either the heavy or light chain. Exemplary additional antigen-binding units include single domain antibodies (e.g., variable heavy chain or variable light chain), engineered protein scaffolds, and paired antibody variable domains (e.g., single chain variable fragments or variable fragments). See Id. Examples of appended IgG formats include dual variable domain IgG (DVD-Ig), IgG(H)-scFv, scFv-(H)IgG, IgG(L)-scFv, scFv-(L)IgG, IgG(L,H)-Fv, IgG(H)-V, V(H)-IgG, IgG(L)-V, V(L)-IgG, KIH IgG-scFab, 2scFv-IgG, IgG-2scFv, scFv4-Ig, zybody, and DVI-IgG (four-in-one). See Spiess et al. Mol. Immunol. 67(2015):95-106. An example of an IgG-scFv is MM-141 (Merrimack Pharmaceuticals), which binds IGF-1R and HER3. Examples of DVD-Ig include ABT-981 (AbbVie), which binds IL-1α and IL-1β; and ABT-122 (AbbVie), which binds TNF and IL-17A.

Bispecific antibody fragments (BsAb) are a format of bispecific antibody molecules that lack some or all of the antibody constant domains. For example, some BsAb lack an Fc region. In embodiments, bispecific antibody fragments include heavy and light chain regions that are connected by a peptide linker that permits efficient expression of the BsAb in a single host cell. Exemplary bispecific antibody fragments include but are not limited to nanobody, nanobody-HAS, BiTE, Diabody, DART, TandAb, scDiabody, scDiabody-CH3, Diabody-CH3, triple body, miniantibody, minibody, TriBi minibody, scFv-CH3 KIH, Fab-scFv, scFv-CH-CL-scFv, F(ab′)2, F(ab′)2-scFv2, scFv-KIH, Fab-scFv-Fc, tetravalent HCAb, scDiabody-Fc, Diabody-Fc, tandem scFv-Fc, and intrabody. See Id. For example, the BiTE format comprises tandem scFvs, where the component scFvs bind to CD3 on T cells and a surface antigen on cancer cells

Bispecific fusion proteins include antibody fragments linked to other proteins, e.g., to add additional specificity and/or functionality. An example of a bispecific fusion protein is an immTAC, which comprises an anti-CD3 scFv linked to an affinity-matured T-cell receptor that recognizes HLA-presented peptides. In embodiments, the dock-and-lock (DNL) method can be used to generate bispecific antibody molecules with higher valency. Also, fusions to albumin binding proteins or human serum albumin can be extend the serum half-life of antibody fragments. See Id.

In embodiments, chemical conjugation, e.g., chemical conjugation of antibodies and/or antibody fragments, can be used to create BsAb molecules. See Id. An exemplary bispecific antibody conjugate includes the CovX-body format, in which a low molecular weight drug is conjugated site-specifically to a single reactive lysine in each Fab arm or an antibody or fragment thereof. In embodiments, the conjugation improves the serum half-life of the low molecular weight drug. An exemplary CovX-body is CVX-241 (NCT01004822), which comprises an antibody conjugated to two short peptides inhibiting either VEGF or Ang2. See Id.

The antibody molecules can be produced by recombinant expression, e.g., of at least one or more component, in a host system. Exemplary host systems include eukaryotic cells (e.g., mammalian cells, e.g., CHO cells, or insect cells, e.g., SF9 or S2 cells) and prokaryotic cells (e.g., E. coli). Bispecific antibody molecules can be produced by separate expression of the components in different host cells and subsequent purification/assembly. Alternatively, the antibody molecules can be produced by expression of the components in a single host cell. Purification of bispecific antibody molecules can be performed by various methods such as affinity chromatography, e.g., using protein A and sequential pH elution. In other embodiments, affinity tags can be used for purification, e.g., histidine-containing tag, myc tag, or streptavidin tag.

Exemplary Bispecific Molecules

In an aspect, a multispecific molecule disclosed herein comprises a sequence disclosed herein, e.g., a sequence chosen from SEQ ID NOs: 1004-1007, 3275-3277, 3286, or 3287, or a sequence with at least 85%, 90%, 955, 96%, 97%, 98%, 99% or more identity thereto. In some embodiments, a multispecific molecule disclosed herein comprises a leader sequence comprising the amino acid sequence of SEQ ID NO: 3288. In some embodiments, a multispecific molecule disclosed herein does not comprise a leader sequence comprising the amino acid sequence of SEQ ID NO: 3288.

Molecule F: aCD19×aVb6.5

Molecule F comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1004 and a light chain comprising the amino acid sequence of SEQ ID NO: 1005.

Molecule F.1 (heavy chain) (Tcrvbeta6_5 scFv/anti-CD19 heavy chain) SEQ ID NO: 1004 METDTLLLWVLLLWVPGSTGQVQLVQSGAEVKKPGSSVKVSCKASGYSFT TYYIHWVRQAPGQGLEWMGWFFPGSGNIKYNEKFKGRVTITADTSTSTAY MELSSLRSEDTAVYYCAGSYYSYDVLDYWGQGTTVTVSSGGGGSGGGGSG GGGSGGGGSDIQMTQSPSFLSASVGDRVTITCKASQNVGINVVWHQQKPG KAPKALIYSSSHRYSGVPSRFSGSGSGTEFTLTISSLQPEDFATYFCQQF KSYPLTFGQGTKLEIKGGGGSQVTLRESGPALVKPTQTLTLTCTFSGFSL STSGMGVGWIRQPPGKALEWLAHIWWDDDKRYNPALKSRLTISKDTSKNQ VFLTMTNMDPVDTATYYCARMELWSYYFDYWGQGTTVTVSSASTKGPSVF PLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQS SGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTC PPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFN WYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNK ALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSD IAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCS VMHEALHNHYTQKSLSLSPGK Molecule F.2 (light chain) (anti-CD19 light chain) SEQ ID NO: 1005 METPAQLLFLLLLWLPDTTGENVLTQSPATLSLSPGERATLSCSASSSVS YMHWYQQKPGQAPRLLIYDTSKLASGIPARFSGSGSGTDHTLTISSLEPE DFAVYYCFQGSVYPFTFGQGTKLEIKRTVAAPSVFIFPPSDEQLKSGTAS VVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTL SKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC

In an aspect, a multispecific molecule disclosed herein comprises SEQ ID NO: 1004 and/or SEQ ID NO: 1005 or a sequence with at least 85%, 90%, 955, 96%, 97%, 98%, 99% or more identity thereto.

Molecule G: aBCMA x aVb6.5

Molecule G comprises a heavy chain comprising the amino acid sequence of SEQ ID NO: 1006 and a light chain comprising the amino acid sequence of SEQ ID NO: 1007.

Molecule G.1 (heavy chain) SEQ ID NO: 1006 METDTLLLWVLLLWVPGSTGQVQLVQSGAEVKKPGSSVKVSCKASGYSFT TYYIHWVRQAPGQGLEWMGWFFPGSGNIKYNEKFKGRVTITADTSTSTAY MELSSLRSEDTAVYYCAGSYYSYDVLDYWGQGTTVTVSSGGGGSGGGGSG GGGSGGGGSDIQMTQSPSFLSASVGDRVTITCKASQNVGINVVWHQQKPG KAPKALIYSSSHRYSGVPSRFSGSGSGTEFTLTISSLQPEDFATYFCQQF KSYPLTFGQGTKLEIKGGGGSQVQLVESGGGVVQPGRSLRLSCAASGIDF SRYWMSWVRQAPGKGLEWVGEINPDSSTINYAPSLKDRFTISRDNSKNTL YLQMSSLRAEDTAVYYCASLYYDYGDAMDYWGQGTTVTVSSASTKGPSVF PLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQS SGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTC PPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFN WYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNK ALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSD IAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCS VMHEALHNRFTQKSLSLSPGK Molecule G.2 (light chain) SEQ ID NO: 1007 METDTLLLWVLLLWVPGSTGDIQMTQSPSSLSASVGDRVTITCKASQSVD SNVAWYQQKPEKAPKALIFSASLRFSGVPSRFSGSGSGTDFTLTISSLQP EDFATYFCQQYNNYPLTFGQGTKLEIKRTVAAPSVFIFPPSDEQLKSGTA SVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLT LSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC

In an aspect, a multispecific molecule disclosed herein comprises SEQ ID NO: 1006 and/or SEQ ID NO: 1007 or a sequence with at least 85%, 90%, 955, 96%, 97%, 98%, 99% or more identity thereto.

Molecule H: aBCMA x aTCRvbeta6_5

Molecule H comprises a first heavy chain comprising the amino acid sequence of SEQ ID NO: 3275, a light chain comprising the amino acid sequence of SEQ ID NO: 3277, and a second heavy chain comprising the amino acid sequence of SEQ ID NO: 3276.

Molecule H.1 (anti-BCMA heavy chain) SEQ ID NO: 3275 METDTLLLWVLLLWVPGSTGQVQLVESGGGVVQPGRSLRLSCAASGIDFS RYWMSWVRQAPGKGLEWVGEINPDSSTINYAPSLKDRFTISRDNSKNTLY LQMSSLRAEDTAVYYCASLYYDYGDAMDYWGQGTTVTVSSASTKGPSVFP LAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSS GLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKRVEPKSCDKTHTCP PCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNW YVDGVEVHNAKTKPREEQYNATYRVVSVLTVLHQDWLNGKEYKCKVSNKA LPAPIEKTISKAKGQPREPQVYTLPPCREEMTKNQVSLWCLVKGFYPSDI AVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSV MHEALHNHYTQKSLSLSPGK Molecule H.2 (TCRvbeta_6_5 scFv humanized) SEQ ID NO: 3276 METDTLLLWVLLLWVPGSTGQVQLVQSGAEVKKPGSSVKVSCKASGYSFT TYYIHWVRQAPGQGLEWMGWFFPGSGNIKYNEKFKGRVTITADTSTSTAY MELSSLRSEDTAVYYCAGSYYSYDVLDYWGQGTTVTVSSGGGGSGGGGSG GGGSGGGGSDIQMTQSPSFLSASVGDRVTITCKASQNVGINVVWHQQKPG KAPKALIYSSSHRYSGVPSRFSGSGSGTEFTLTISSLQPEDFATYFCQQF KSYPLTFGQGTKLEIKGGGGSGGGGSDKTHTCPPCPAPELLGGPSVFLFP PKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREE QYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPR EPQVCTLPPSREEMTKNQVSLSCAVKGFYPSDIAVEWESNGQPENNYKTT PPVLDSDGSFFLVSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLS PGK Molecule H.3 (anti-BCMA light chain) SEQ ID NO: 3277 METDTLLLWVLLLWVPGSTGDIQMTQSPSSLSASVGDRVTITCKAS QSVDSNVAWYQQKPEKAPKALIFSASLRFSGVPSRFSGSGSGTDFTLTIS SLQPEDFATYFCQQYNNYPLTFGQGTKLEIKRTVAAPSVFIFPPSDEQLK SGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLS STLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC

In an aspect, a multispecific molecule disclosed herein comprises SEQ ID NO: 3275, SEQ ID NO: 3276, and/or SEQ ID NO: 3277 or a sequence with at least 85%, 90%, 955, 96%, 97%, 98%, 99% or more identity thereto.

Molecule I: Half Arm BCMA Fab with c-Terminal scFv TCRvbeta

Molecule I comprises a first heavy chain comprising the amino acid sequence of SEQ ID NO: 3286, a light chain comprising the amino acid sequence of SEQ ID NO: 3277, and a second heavy chain comprising the amino acid sequence of SEQ ID NO: 3287.

Molecule I.1 (heavy chain 1) SEQ ID NO: 3286 METDTLLLWVLLLWVPGSTGQVQLVESGGGVVQPGRSLRLSCAASGIDFS RYWMSWVRQAPGKGLEWVGEINPDSSTINYAPSLKDRFTISRDNSKNTLY LQMSSLRAEDTAVYYCASLYYDYGDAMDYWGQGTTVTVSSASTKGPSVFP LAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSS GLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKRVEPKSCDKTHTCP PCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNW YVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKA LPAPIEKTISKAKGQPREPQVYTLPPCREEMTKNQVSLWCLVKGFYPSDI AVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSV MHEALHNHYTQKSLSLSPGKGGGGSGGGGSGGGGSEVQLVESGGGLVQPG GSLRLSCAASGFTFSTYAMNWVRQAPGKGLEWVSRIRSKYNNYATYYADS VKDRFTISRDDSKNTLYLQMNSLKTEDTAVYYCVRHGNFGNSYVSWFAYW GQGTLVTVSSGGGGSGGGGSGGGGSGGGGSQAVVTQEPSLTVSPGGTVTL TCRSSTGAVTTSNYANWVQQKPGQAPRGLIGGTNKRAPWTPARFSGSLLG GKAALTLSGAQPEDEAEYYCALWYSNLWVFGGGTKLTVL Molecule I.2 (light chain) SEQ ID NO: 3277 METDTLLLWVLLLWVPGSTGDIQMTQSPSSLSASVGDRVTITCKASQSVD SNVAWYQQKPEKAPKALIFSASLRFSGVPSRFSGSGSGTDFTLTISSLQP EDFATYFCQQYNNYPLTFGQGTKLEIKRTVAAPSVFIFPPSDEQLKSGTA SVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLT LSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC Molecule I.3 (heavy chain 2) SEQ ID NO: 3287 METDTLLLWVLLLWVPGSTGDKTHTCPPCPAPELLGGPSVFLFPPKPKDT LMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTY RVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVCT LPPSREEMTKNQVSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDS DGSFFLVSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK

In an aspect, a multispecific molecule disclosed herein comprises SEQ ID NO: 3286, SEQ ID NO: 3277, and/or SEQ ID NO: 3287 or a sequence with at least 85%, 90%, 955, 96%, 97%, 98%, 99% or more identity thereto.

Antibody-Like Frameworks or Scaffolds

A wide variety of antibody/immunoglobulin frameworks or scaffolds can be employed in the anti-TCRvb antibody molecules disclosed herein or multifunctional formats thereof so long as the resulting polypeptide includes at least one binding region which specifically binds to the target antigen, e.g., a TCRvb, a tumor antigen, among others. Such frameworks or scaffolds include the 5 main idiotypes of human immunoglobulins, or fragments thereof, and include immunoglobulins of other animal species, preferably having humanized aspects. Novel frameworks, scaffolds and fragments continue to be discovered and developed by those skilled in the art.

In one embodiment, the anti-TCRvb antibody molecules disclosed herein or multifunctional formats thereof include non-immunoglobulin based antibodies using non-immunoglobulin scaffolds onto which CDRs can be grafted. Any non-immunoglobulin frameworks and scaffolds may be employed, as long as they comprise a binding region specific for the target antigen (e.g., TCRvb or a tumor antigen). Exemplary non-immunoglobulin frameworks or scaffolds include, but are not limited to, fibronectin (Compound Therapeutics, Inc., Waltham, MA), ankyrin (Molecular Partners AG, Zurich, Switzerland), domain antibodies (Domantis, Ltd., Cambridge, MA, and Ablynx nv, Zwijnaarde, Belgium), lipocalin (Pieris Proteolab AG, Freising, Germany), small modular immuno-pharmaceuticals (Trubion Pharmaceuticals Inc., Seattle, WA), maxybodies (Avidia, Inc., Mountain View, CA), Protein A (Affibody AG, Sweden), and affilin (gamma-crystallin or ubiquitin) (Scil Proteins GmbH, Halle, Germany).

Fibronectin scaffolds are typically based on fibronectin type III domain (e.g., the tenth module of the fibronectin type III (10 Fn3 domain)). The fibronectin type III domain has 7 or 8 beta strands which are distributed between two beta sheets, which themselves pack against each other to form the core of the protein, and further containing loops (analogous to CDRs) which connect the beta strands to each other and are solvent exposed. There are at least three such loops at each edge of the beta sheet sandwich, where the edge is the boundary of the protein perpendicular to the direction of the beta strands (see U.S. Pat. No. 6,818,418). Because of this structure, the non-immunoglobulin antibody mimics antigen binding properties that are similar in nature and affinity to those of antibodies. These scaffolds can be used in a loop randomization and shuffling strategy in vitro that is similar to the process of affinity maturation of antibodies in vivo. These fibronectin-based molecules can be used as scaffolds where the loop regions of the molecule can be replaced with CDRs of the invention using standard cloning techniques.

The ankyrin technology is based on using proteins with ankyrin derived repeat modules as scaffolds for bearing variable regions which can be used for binding to different targets. The ankyrin repeat module typically is a about 33 amino acid polypeptide consisting of two anti-parallel α-helices and a β-turn. Binding of the variable regions can be optimized by using ribosome display.

Avimers are used by nature for protein-protein interactions and in human over 250 proteins are structurally based on A-domains. Avimers consist of a number of different “A-domain” monomers (2-10) linked via amino acid linkers. Avimers can be created that can bind to the target antigen using the methodology described in, for example, U.S. Patent Application Publication Nos. 20040175756; 20050053973; 20050048512; and 20060008844.

Affibody affinity ligands are small, simple proteins composed of a three-helix bundle based on the scaffold of one of the IgG-binding domains of Protein A. Protein A is a surface protein from the bacterium Staphylococcus aureus. This scaffold domain consists of 58 amino acids, 13 of which are randomized to generate affibody libraries with a large number of ligand variants (See e.g., U.S. Pat. No. 5,831,012). Affibody molecules mimic antibodies, they have a molecular weight of 6 kDa, compared to the molecular weight of antibodies, which is 150 kDa. In spite of its small size, the binding site of affibody molecules is similar to that of an antibody.

Anticalins are known commercially, e.g., Pieris ProteoLab AG. They are derived from lipocalins, a widespread group of small and robust proteins that are usually involved in the physiological transport or storage of chemically sensitive or insoluble compounds. Several natural lipocalins occur in human tissues or body liquids. The protein architecture is reminiscent of immunoglobulins, with hypervariable loops on top of a rigid framework. However, in contrast with antibodies or their recombinant fragments, lipocalins are composed of a single polypeptide chain with 160 to 180 amino acid residues, being just marginally bigger than a single immunoglobulin domain. The set of four loops, which makes up the binding pocket, shows pronounced structural plasticity and tolerates a variety of side chains. The binding site can thus be reshaped in a proprietary process in order to recognize prescribed target molecules of different shape with high affinity and specificity. One protein of lipocalin family, the bilin-binding protein (BBP) of Pieris Brassicae has been used to develop anticalins by mutagenizing the set of four loops. One example of a patent application describing anticalins is in PCT Publication No. WO 199916873.

Affilin molecules are small non-immunoglobulin proteins which are designed for specific affinities towards proteins and small molecules. New affilin molecules can be very quickly selected from two libraries, each of which is based on a different human derived scaffold protein. Affilin molecules do not show any structural homology to immunoglobulin proteins. Currently, two affilin scaffolds are employed, one of which is gamma crystalline, a human structural eye lens protein and the other is “ubiquitin” superfamily proteins. Both human scaffolds are very small, show high temperature stability and are almost resistant to pH changes and denaturing agents. This high stability is mainly due to the expanded beta sheet structure of the proteins. Examples of gamma crystalline derived proteins are described in WO200104144 and examples of “ubiquitin-like” proteins are described in WO2004106368.

Protein epitope mimetics (PEM) are medium-sized, cyclic, peptide-like molecules (MW 1-2 kDa) mimicking beta-hairpin secondary structures of proteins, the major secondary structure involved in protein-protein interactions.

Domain antibodies (dAbs) can be used in the anti-TCRvb antibody molecules disclosed herein or multifunctional formats thereof are small functional binding fragments of antibodies, corresponding to the variable regions of either the heavy or light chains of antibodies. Domain antibodies are well expressed in bacterial, yeast, and mammalian cell systems. Further details of domain antibodies and methods of production thereof are known in the art (see, for example, U.S. Pat. Nos. 6,291,158; 6,582,915; 6,593,081; 6,172,197; 6,696,245; European Patents 0368684 & 0616640; WO05/035572, WO04/101790, WO04/081026, WO04/058821, WO04/003019 and WO03/002609. Nanobodies are derived from the heavy chains of an antibody.

A nanobody typically comprises a single variable domain and two constant domains (CH2 and CH3) and retains antigen-binding capacity of the original antibody. Nanobodies can be prepared by methods known in the art (See e.g., U.S. Pat. Nos. 6,765,087, 6,838,254, WO 06/079372). Unibodies consist of one light chain and one heavy chain of an IgG4 antibody. Unibodies may be made by the removal of the hinge region of IgG4 antibodies. Further details of unibodies and methods of preparing them may be found in WO2007/059782.

Tumor Antigen Moiety

In an aspect, provided herein is a multispecific molecule, e.g., a bispecific molecule, comprising:

    • (i) a first moiety (e.g., a first immune cell engager) comprising the anti-TCRβV antibody molecule described herein; and
    • (ii) a second moiety comprising one or more of: a tumor-targeting moiety; a second immune cell engager; a cytokine molecule or a stromal modifying moiety.

In some embodiments of any of the compositions or methods disclosed herein, the tumor-targeting moiety is an antigen, e.g., a cancer antigen. In some embodiments, the cancer antigen is a tumor antigen or stromal antigen, or a hematological antigen.

In some embodiments of any of the compositions or methods disclosed herein, the tumor-targeting moiety, e.g., cancer antigen, is chosen from: BCMA, FcRH5, CD19, CD20, CD22, CD30, CD33, CD38, CD47, CD99, CD123, FcRH5, CLEC12, CD179A, SLAMF7, or NY-ESO1, PDL1, CD47, gangloside 2 (GD2), prostate stem cell antigen (PSCA), prostate specific membrane antigen (PMSA), prostate-specific antigen (PSA), carcinoembryonic antigen (CEA), Ron Kinase, c-Met, Immature laminin receptor, TAG-72, BING-4, Calcium-activated chloride channel 2, Cyclin-B1, 9D7, Ep-CAM, EphA3, Her2/neu, Telomerase, SAP-1, Survivin, NY-ESO-1/LAGE-1, PRAME, SSX-2, Melan-A/MART-1, Gp100/pme117, Tyrosinase, TRP-1/-2, MC1R, β-catenin, BRCA1/2, CDK4, CML66, Fibronectin, p53, Ras, TGF-B receptor, AFP, ETA, MAGE, MUC-1, CA-125, BAGE, GAGE, NY-ESO-1, β-catenin, CDK4, CDC27, α actinin-4, TRP1/gp75, TRP2, gp100, Melan-A/MART1, gangliosides, WT1, EphA3, Epidermal growth factor receptor (EGFR), MART-2, MART-1, MUC1, MUC2, MUM1, MUM2, MUM3, NA88-1, NPM, OA1, OGT, RCC, RUI1, RUI2, SAGE, TRG, TRP1, TSTA, Folate receptor alpha, L1-CAM, CAIX, gpA33, GD3, GM2, VEGFR, Integrins (Integrin alphaVbeta3, Integrin alpha5Beta1), Carbohydrates (Le), IGF1R, EPHA3, TRAILR1, TRAILR2, RANKL, (FAP), TGF-beta, hyaluronic acid, collagen, e.g., collagen IV, tenascin C, or tenascin W. In some embodiments, the tumor-targeting moiety, e.g., cancer antigen, is BCMA. In some embodiments, the tumor-targeting moiety, e.g., cancer antigen, is FcRH5.

In some embodiments of any of the compositions or methods disclosed herein, the tumor-targeting moiety, e.g., cancer antigen, is chosen from: CD19, CD123, CD22, CD30, CD171, CS-1, C-type lectin-like molecule-1, CD33, epidermal growth factor receptor variant III (EGFRvIII), ganglioside G2 (GD2), ganglioside GD3, TNF receptor family member B cell maturation (BCMA), Tn antigen ((Tn Ag) or (GalNAcα-Ser/Thr)), prostate-specific membrane antigen (PSMA), Receptor tyrosine kinase-like orphan receptor 1 (ROR1), Fms-Like Tyrosine Kinase 3 (FLT3), Tumor-associated glycoprotein 72 (TAG72), CD38, CD44v6, Carcinoembryonic antigen (CEA), Epithelial cell adhesion molecule (EPCAM), B7H3 (CD276), KIT (CD117), Interleukin-13 receptor subunit alpha-2, mesothelin, Interleukin 11 receptor alpha (IL-11Ra), prostate stem cell antigen (PSCA), Protease Serine 21, vascular endothelial growth factor receptor 2 (VEGFR2), Lewis(Y) antigen, CD24, Platelet-derived growth factor receptor beta (PDGFR-beta), Stage-specific embryonic antigen-4 (SSEA-4), CD20, Folate receptor alpha, Receptor tyrosine-protein kinase ERBB2 (Her2/neu), Mucin 1, cell surface associated (MUC1), epidermal growth factor receptor (EGFR), neural cell adhesion molecule (NCAM), Prostase, prostatic acid phosphatase (PAP), elongation factor 2 mutated (ELF2M), Ephrin B2, fibroblast activation protein alpha (FAP), insulin-like growth factor 1 receptor (IGF-I receptor), carbonic anhydrase IX (CAIX), Proteasome (Prosome, Macropain) Subunit, Beta Type, 9 (LMP2), glycoprotein 100 (gp100), oncogene fusion protein consisting of breakpoint cluster region (BCR) and Abelson murine leukemia viral oncogene homolog 1 (Abl) (bcr-abl), tyrosinase, ephrin type-A receptor 2 (EphA2), Fucosyl GM1, sialyl Lewis adhesion molecule (sLe), ganglioside GM3, transglutaminase 5 (TGS5), high molecular weight-melanoma-associated antigen (HMWMAA), o-acetyl-GD2 ganglioside (OAcGD2), Folate receptor beta, tumor endothelial marker 1 (TEM1/CD248), tumor endothelial marker 7-related (TEM7R), claudin 6 (CLDN6), thyroid stimulating hormone receptor (TSHR), G protein-coupled receptor class C group 5, member D (GPRC5D), chromosome X open reading frame 61 (CXORF61), CD97, CD179a, anaplastic lymphoma kinase (ALK), Polysialic acid, placenta-specific 1 (PLAC1), hexasaccharide portion of globoH glycoceramide (GloboH), mammary gland differentiation antigen (NY-BR-1), uroplakin 2 (UPK2), Hepatitis A virus cellular receptor 1 (HAVCR1), adrenoceptor beta 3 (ADRB3), pannexin 3 (PANX3), G protein-coupled receptor 20 (GPR20), lymphocyte antigen 6 complex, locus K 9 (LY6K), Olfactory receptor 51E2 (OR51E2), TCR Gamma Alternate Reading Frame Protein (TARP), Wilms tumor protein (WT1), Cancer/testis antigen 1 (NY-ESO-1), Cancer/testis antigen 2 (LAGE-1a), Melanoma-associated antigen 1 (MAGE-A1), ETS translocation-variant gene 6, located on chromosome 12p (ETV6-AML), sperm protein 17 (SPA17), X Antigen Family, Member 1A (XAGE1), angiopoietin-binding cell surface receptor 2 (Tie 2), melanoma cancer testis antigen-1 (MAD-CT-1), melanoma cancer testis antigen-2 (MAD-CT-2), Fos-related antigen 1, tumor protein p53 (p53), p53 mutant, prostein, surviving, telomerase, prostate carcinoma tumor antigen-1, melanoma antigen recognized by T cells 1, Rat sarcoma (Ras) mutant, human Telomerase reverse transcriptase (hTERT), sarcoma translocation breakpoints, melanoma inhibitor of apoptosis (ML-IAP), ERG (transmembrane protease, serine 2 (TMPRSS2) ETS fusion gene), N-Acetyl glucosaminyl-transferase V (NA17), paired box protein Pax-3 (PAX3), Androgen receptor, Cyclin B1, v-myc avian myelocytomatosis viral oncogene neuroblastoma derived homolog (MYCN), Ras Homolog Family Member C (RhoC), Tyrosinase-related protein 2 (TRP-2), Cytochrome P450 1B1 (CYP1B1), CCCTC-Binding Factor (Zinc Finger Protein)-Like, Squamous Cell Carcinoma Antigen Recognized By T Cells 3 (SART3), Paired box protein Pax-5 (PAX5), proacrosin binding protein sp32 (OY-TES1), lymphocyte-specific protein tyrosine kinase (LCK), A kinase anchor protein 4 (AKAP-4), synovial sarcoma, X breakpoint 2 (SSX2), Receptor for Advanced Glycation Endproducts (RAGE-1), renal ubiquitous 1 (RU1), renal ubiquitous 2 (RU2), legumain, human papilloma virus E6 (HPV E6), human papilloma virus E7 (HPV E7), intestinal carboxyl esterase, heat shock protein 70-2 mutated (mut hsp70-2), CD79a, CD79b, CD72, Leukocyte-associated immunoglobulin-like receptor 1 (LAIR1), Fc fragment of IgA receptor (FCAR or CD89), Leukocyte immunoglobulin-like receptor subfamily A member 2 (LILRA2), CD300 molecule-like family member f (CD300LF), C-type lectin domain family 12 member A (CLEC12A), bone marrow stromal cell antigen 2 (BST2), EGF-like module-containing mucin-like hormone receptor-like 2 (EMR2), lymphocyte antigen 75 (LY75), Glypican-3 (GPC3), Fc receptor-like 5 (FCRL5), or immunoglobulin lambda-like polypeptide 1 (IGLL1).

FcRH5 Targeting Moieties

In some embodiments, the multispecific molecules disclosed herein include a targeting moiety that binds to FcRH5 (e.g., a FcRH5 targeting moiety). The FcRH5 targeting moiety can be chosen from an antibody molecule (e.g., an antigen binding domain as described herein), a receptor or a receptor fragment, or a ligand or a ligand fragment, or a combination thereof. In some embodiments, the FcRH5 targeting moiety associates with, e.g., binds to, a cancer or hematopoietic cell (e.g., a molecule, e.g., antigen, present on the surface of the cancer or hematopoietic cell). In certain embodiments, the FcRH5 targeting moiety targets, e.g., directs the multispecific molecules disclosed herein to a cancer or hematopoietic cell. In some embodiments, the cancer is a hematological cancer, e.g., multiple myeloma.

In some embodiments, the multispecific molecule, e.g., the FcRH5 targeting moiety, binds to a FcRH5 antigen on the surface of a cell, e.g., a cancer or hematopoietic cell. The FcRH5 antigen can be present on a primary tumor cell, or a metastatic lesion thereof. In some embodiments, the cancer is a hematological cancer, e.g., multiple myeloma. For example, the FcRH5 antigen can be present on a tumor, e.g., a tumor of a class typified by having one or more of: limited tumor perfusion, compressed blood vessels, or fibrotic tumor interstitium.

The multispecific molecules described herein includes a FcRH5 targeting moiety that comprises an anti-FcRH5 antibody or antigen-binding fragment thereof described in U.S. Pat. No. 7,999,077, US20150098900, U.S. Pat. Nos. 8,299,220, 7,105,149, 8,362,213, 8,466,260, 8,617,559, US20160368985, US20150166661, and US20080247944, the entire contents of any of the aforesaid publications are herein incorporated by reference.

In some embodiments, the multispecific molecules described herein includes a FcRH5 targeting moiety that comprises an anti-FcRH5 antibody or antigen-binding fragment thereof described in U.S. Pat. No. 7,999,077, the entire contents of which are herein incorporated by reference.

BCMA Targeting Moieties

In certain embodiments, the multispecific molecules disclosed herein include a targeting moiety that binds to BCMA (e.g., a BCMA targeting moiety). The BCMA targeting moiety can be chosen from an antibody molecule (e.g., an antigen binding domain as described herein), a receptor or a receptor fragment, or a ligand or a ligand fragment, or a combination thereof. In some embodiments, the BCMA targeting moiety associates with, e.g., binds to, a cancer or hematopoietic cell (e.g., a molecule, e.g., antigen, present on the surface of the cancer or hematopoietic cell). In certain embodiments, the BCMA targeting moiety targets, e.g., directs the multispecific molecules disclosed herein to a cancer or hematopoietic cell. In some embodiments, the cancer is a hematological cancer, e.g., multiple myeloma.

In some embodiments, the multispecific molecule, e.g., the BCMA targeting moiety, binds to a BCMA antigen on the surface of a cell, e.g., a cancer or hematopoietic cell. The BCMA antigen can be present on a primary tumor cell, or a metastatic lesion thereof. In some embodiments, the cancer is a hematological cancer, e.g., multiple myeloma. For example, the BCMA antigen can be present on a tumor, e.g., a tumor of a class typified by having one or more of: limited tumor perfusion, compressed blood vessels, or fibrotic tumor interstitium.

Exemplary BCMA Targeting Moieties

The multispecific molecules described herein can include a BCMA targeting moiety that comprises an anti-BCMA antibody or antigen-binding fragment thereof described in U.S. Pat. Nos. 8,920,776, 9,243,058, 9,340,621, 8,846,042, 7,083,785, 9,545,086, 7,276,241, 9,034,324, 7,799,902, 9,387,237, 8,821,883, 861,745, US20130273055, US20160176973, US20150368351, US20150376287, US20170022284, US20160015749, US20140242077, US20170037128, US20170051068, US20160368988, US20160311915, US20160131654, US20120213768, US20110177093, US20160297885, EP3137500, EP2699259, EP2982694, EP3029068, EP3023437, WO2016090327, WO2017021450, WO2016110584, WO2016118641, WO2016168149, the entire contents of which are incorporated herein by reference.

In one embodiment, the BCMA-targeting moiety includes an antibody molecule (e.g., Fab or scFv) that binds to BCMA. In some embodiments, the antibody molecule to BCMA comprises one, two, or three CDRs from any of the heavy chain variable domain sequences of Table 1, or a closely related CDR, e.g., CDRs which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) from any of the CDR sequences of Table 14. In some embodiments, the antibody molecule to BCMA comprises a heavy chain variable domain sequence chosen from any of the amino acid sequences of Table 14, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions)).

Alternatively, or in combination with the heavy chain to BCMA disclosed herein, the antibody molecule to BCMA comprises one, two, or three CDRs from any of the light chain variable domain sequences of Table 14, or a closely related CDR, e.g., CDRs which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) from any of the CDR sequences of Table 14. In some embodiments, the antibody molecule to BCMA comprises a light chain variable domain sequence chosen from any of the amino acid sequences of Table 14, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions)).

TABLE 9 Amino acid sequences of exemplary variable regions of anti-BCMA antibodies. SEQ ID NO Description Sequence 3439 83A10 VH EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGL EWVSAISGSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAED TAVYYCAKVLGWFDYWGQGTLVTVSS 3440 83A10 VL EIVLTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQKPGQAPR LLIYGASSRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQYG YPPDFTFGQGTKVEIK 3441 17A5 VH EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGL EWVSAISGSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAED TAVYYCAKVAPYFAPFDYWGQGTLVTVSS 3442 17A5 VL EIVLTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQKPGQAPR LLIYGASSRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQYG NPPLYTFGQGTKVEIK 3443 13A4 VH EVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGL EWMGIIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDT AMYYCARNGYLGDYWGQGTLVTVSS 3444 13A4 VL DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPG QSPQLLIYLGSNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYY CMQAMQIPTFGQGTKVEIK 3445 J22.9-xi VH QVQLQQSGGGLVQPGGSLKLSCAASGIDFSRYWMSWVRRAPGKG LEWIGEINPDSSTINYAPSLKDKFIISRDNAKNTLYLQMSKVRSEDT ALYYCASLYYDYGDAMDYWGQGTSVTVSS 3446 J22.9-xi VL DIVMTQSQRFMTTSVGDRVSVTCKASQSVDSNVAWYQQKPRQSP KALIFSASLRFSGVPARFTGSGSGTDFTLTISNLQSEDLAEYFCQQY NNYPLTFGAGTKLELKR 3447 2A1 VH EVQLVESGGGLVKPGGSLRLSCAASGFTFGDYALSWFRQAPGKGL EWVGVSRSKAYGGTTDYAASVKGRFTISRDDSKSTAYLQMNSLKT EDTAVYYCASSGYSSGWTPFDYWGQGTLVTVSS 3448 2A1 VL QSVLTQPPSASGTPGQRVTISCSGSSSNIGSNTVNWYQQLPGTAPKL LIFNYHQRPSGVPDRFSGSKSGSSASLAISGLQSEDEADYYCAAWD DSLNGWVFGGGTKLTVLG

CDR-Grafted Scaffolds

In embodiments, the antibody molecule is a CDR-grafted scaffold domain. In embodiments, the scaffold domain is based on a fibronectin domain, e.g., fibronectin type III domain. The overall fold of the fibronectin type III (Fn3) domain is closely related to that of the smallest functional antibody fragment, the variable domain of the antibody heavy chain. There are three loops at the end of Fn3; the positions of BC, DE and FG loops approximately correspond to those of CDR1, 2 and 3 of the VH domain of an antibody. Fn3 does not have disulfide bonds; and therefore Fn3 is stable under reducing conditions, unlike antibodies and their fragments (see, e.g., WO 98/56915; WO 01/64942; WO 00/34784). An Fn3 domain can be modified (e.g., using CDRs or hypervariable loops described herein) or varied, e.g., to select domains that bind to an antigen/marker/cell described herein.

In embodiments, a scaffold domain, e.g., a folded domain, is based on an antibody, e.g., a “minibody” scaffold created by deleting three beta strands from a heavy chain variable domain of a monoclonal antibody (see, e.g., Tramontano et al., 1994, J Mol. Recognit. 7:9; and Martin et al., 1994, EMBO J. 13:5303-5309). The “minibody” can be used to present two hypervariable loops. In embodiments, the scaffold domain is a V-like domain (see, e.g., Coia et al. WO 99/45110) or a domain derived from tendamistatin, which is a 74 residue, six-strand beta sheet sandwich held together by two disulfide bonds (see, e.g., McConnell and Hoess, 1995, J Mol. Biol. 250:460). For example, the loops of tendamistatin can be modified (e.g., using CDRs or hypervariable loops) or varied, e.g., to select domains that bind to a marker/antigen/cell described herein. Another exemplary scaffold domain is a beta-sandwich structure derived from the extracellular domain of CTLA-4 (see, e.g., WO 00/60070).

Other exemplary scaffold domains include but are not limited to T-cell receptors; MHC proteins; extracellular domains (e.g., fibronectin Type III repeats, EGF repeats); protease inhibitors (e.g., Kunitz domains, ecotin, BPTI, and so forth); TPR repeats; trifoil structures; zinc finger domains; DNA-binding proteins; particularly monomeric DNA binding proteins; RNA binding proteins; enzymes, e.g., proteases (particularly inactivated proteases), RNase; chaperones, e.g., thioredoxin, and heat shock proteins; and intracellular signaling domains (such as SH2 and SH3 domains). See, e.g., US 20040009530 and U.S. Pat. No. 7,501,121, incorporated herein by reference.

In embodiments, a scaffold domain is evaluated and chosen, e.g., by one or more of the following criteria: (1) amino acid sequence, (2) sequences of several homologous domains, (3) 3-dimensional structure, and/or (4) stability data over a range of pH, temperature, salinity, organic solvent, oxidant concentration. In embodiments, the scaffold domain is a small, stable protein domain, e.g., a protein of less than 100, 70, 50, 40 or 30 amino acids. The domain may include one or more disulfide bonds or may chelate a metal, e.g., zinc.

Antibody Based Fusions

A variety of formats can be generated which contain additional binding entities attached to the N or C terminus of antibodies. These fusions with single chain or disulfide stabilized Fvs or Fabs result in the generation of tetravalent molecules with bivalent binding specificity for each antigen. Combinations of scFvs and scFabs with IgGs enable the production of molecules which can recognize three or more different antigens.

Antibody Fab Fusion

Antibody-Fab fusions are bispecific antibodies comprising a traditional antibody to a first target and a Fab to a second target fused to the C terminus of the antibody heavy chain. Commonly the antibody and the Fab will have a common light chain. Antibody fusions can be produced by (1) engineering the DNA sequence of the target fusion, and (2) transfecting the target DNA into a suitable host cell to express the fusion protein. It seems like the antibody-scFv fusion may be linked by a (Gly)-Ser linker between the C-terminus of the CH3 domain and the N-terminus of the scFv, as described by Coloma, J. et al. (1997) Nature Biotech 15:159.

Antibody-scFv Fusion

Antibody-scFv Fusions are bispecific antibodies comprising a traditional antibody and a scFv of unique specificity fused to the C terminus of the antibody heavy chain. The scFv can be fused to the C terminus through the Heavy Chain of the scFv either directly or through a linker peptide. Antibody fusions can be produced by (1) engineering the DNA sequence of the target fusion, and (2) transfecting the target DNA into a suitable host cell to express the fusion protein. It seems like the antibody-scFv fusion may be linked by a (Gly)-Ser linker between the C-terminus of the CH3 domain and the N-terminus of the scFv, as described by Coloma, J. et al. (1997) Nature Biotech 15:159.

Variable Domain Immunoglobulin DVD

A related format is the dual variable domain immunoglobulin (DVD), which are composed of VH and VL domains of a second specificity place upon the N termini of the V domains by shorter linker sequences.

Other exemplary multispecific antibody formats include, e.g., those described in the following US20160114057A1, US20130243775A1, US20140051833, US20130022601, US20150017187A1, US20120201746A1, US20150133638A1, US20130266568A1, US20160145340A1, WO2015127158A1, US20150203591A1, US20140322221A1, US20130303396A1, US20110293613, US20130017200A1, US20160102135A1, WO2015197598A2, WO2015197582A1, U.S. Pat. No. 9,359,437, US20150018529, WO2016115274A1, WO2016087416A1, US20080069820A1, U.S. Pat. Nos. 9,145,588B, 7,919,257, and US20150232560A1. Exemplary multispecific molecules utilizing a full antibody-Fab/scFab format include those described in the following, U.S. Pat. No. 9,382,323B2, US20140072581A1, US20140308285A1, US20130165638A1, US20130267686A1, US20140377269A1, U.S. Pat. No. 7,741,446B2, and WO1995009917A1. Exemplary multispecific molecules utilizing a domain exchange format include those described in the following, US20150315296A1, WO2016087650A1, US20160075785A1, WO2016016299A1, US20160130347A1, US20150166670, U.S. Pat. No. 8,703,132B2, US20100316645, U.S. Pat. No. 8,227,577B2, US20130078249.

Fc-Containing Entities (Mini-Antibodies)

Fc-containing entities, also known as mini-antibodies, can be generated by fusing scFv to the C-termini of constant heavy region domain 3 (CH3-scFv) and/or to the hinge region (scFv-hinge-Fc) of an antibody with a different specificity. Trivalent entities can also be made which have disulfide stabilized variable domains (without peptide linker) fused to the C-terminus of CH3 domains of IgGs.

Fc-Containing Multispecific Molecules

In some embodiments, the multispecific molecules disclosed herein includes an immunoglobulin constant region (e.g., an Fc region). Exemplary Fc regions can be chosen from the heavy chain constant regions of IgG1, IgG2, IgG3 or IgG4; more particularly, the heavy chain constant region of human IgG1, IgG2, IgG3, or IgG4.

In some embodiments, the immunoglobulin chain constant region (e.g., the Fc region) is altered, e.g., mutated, to increase or decrease one or more of: Fc receptor binding, antibody glycosylation, the number of cysteine residues, effector cell function, or complement function.

In other embodiments, an interface of a first and second immunoglobulin chain constant regions (e.g., a first and a second Fc region) is altered, e.g., mutated, to increase or decrease dimerization, e.g., relative to a non-engineered interface, e.g., a naturally-occurring interface. For example, dimerization of the immunoglobulin chain constant region (e.g., the Fc region) can be enhanced by providing an Fc interface of a first and a second Fc region with one or more of: a paired protuberance-cavity (“knob-in-a hole”), an electrostatic interaction, or a strand-exchange, such that a greater ratio of heteromultimer to homomultimer forms, e.g., relative to a non-engineered interface.

In some embodiments, the multispecific molecules include a paired amino acid substitution at a position chosen from one or more of 347, 349, 350, 351, 366, 368, 370, 392, 394, 395, 397, 398, 399, 405, 407, or 409, e.g., of the Fc region of human IgG1 For example, the immunoglobulin chain constant region (e.g., Fc region) can include a paired an amino acid substitution chosen from: T366S, L368A, or Y407V (e.g., corresponding to a cavity or hole), and T366W (e.g., corresponding to a protuberance or knob).

In other embodiments, the multifunctional molecule includes a half-life extender, e.g., a human serum albumin or an antibody molecule to human serum albumin.

Heterodimerized Antibody Molecules & Methods of Making

Various methods of producing multispecific antibodies have been disclosed to address the problem of incorrect heavy chain pairing. Exemplary methods are described below. Exemplary multispecific antibody formats and methods of making said multispecific antibodies are also disclosed in e.g., Speiss et al. Molecular Immunology 67 (2015) 95-106; and Klein et al mAbs 4:6, 653-663; November/December 2012; the entire contents of each of which are incorporated by reference herein.

Heterodimerized bispecific antibodies are based on the natural IgG structure, wherein the two binding arms recognize different antigens. IgG derived formats that enable defined monovalent (and simultaneous) antigen binding are generated by forced heavy chain heterodimerization, combined with technologies that minimize light chain mispairing (e.g., common light chain). Forced heavy chain heterodimerization can be obtained using, e.g., knob-in-hole OR strand exchange engineered domains (SEED).

Knob-In-Hole

Knob-in-Hole as described in U.S. Pat. Nos. 5,731,116, 7,476,724 and Ridgway, J. et al. (1996) Prot. Engineering 9(7): 617-621, broadly involves: (1) mutating the CH3 domain of one or both antibodies to promote heterodimerization; and (2) combining the mutated antibodies under conditions that promote heterodimerization. “Knobs” or “protuberances” are typically created by replacing a small amino acid in a parental antibody with a larger amino acid (e.g., T366Y or T366W); “Holes” or “cavities” are created by replacing a larger residue in a parental antibody with a smaller amino acid (e.g., Y407T, T366S, L368A and/or Y407V).

For bispecific antibodies including an Fc domain, introduction of specific mutations into the constant region of the heavy chains to promote the correct heterodimerization of the Fc portion can be utilized. Several such techniques are reviewed in Klein et al. (mAbs (2012) 4:6, 1-11), the contents of which are incorporated herein by reference in their entirety. These techniques include the “knobs-into-holes” (KiH) approach which involves the introduction of a bulky residue into one of the CH3 domains of one of the antibody heavy chains. This bulky residue fits into a complementary “hole” in the other CH3 domain of the paired heavy chain so as to promote correct pairing of heavy chains (see e.g., U.S. Pat. No. 7,642,228).

Exemplary KiH mutations include S354C, T366W in the “knob” heavy chain and Y349C, T366S, L368A, Y407V in the “hole” heavy chain. Other exemplary KiH mutations are provided in Table 4, with additional optional stabilizing Fc cysteine mutations.

TABLE 4 Exemplary Fc KiH mutations and optional Cysteine mutations Position Knob Mutation Hole Mutation T366 T366W T366S L368 L368A Y407 Y407V Additional Cysteine Mutations to form a stabilizing disulfide bridge Position Knob CH3 Hole CH3 S354 S354C Y349 Y349C

Other Fc mutations are provided by Igawa and Tsunoda who identified 3 negatively charged residues in the CH3 domain of one chain that pair with three positively charged residues in the CH3 domain of the other chain. These specific charged residue pairs are: E356-K439, E357-K370, D399-K409 and vice versa. By introducing at least two of the following three mutations in chain A: E356K, E357K and D399K, as well as K370E, K409D, K439E in chain B, alone or in combination with newly identified disulfide bridges, they were able to favor very efficient heterodimerization while suppressing homodimerization at the same time (Martens T et al. A novel one-armed antic-Met antibody inhibits glioblastoma growth in vivo. Clin Cancer Res 2006; 12:6144-52; PMID:17062691). Xencor defined 41 variant pairs based on combining structural calculations and sequence information that were subsequently screened for maximal heterodimerization, defining the combination of S364H, F405A (HA) on chain A and Y349T, T394F on chain B (TF) (Moore G L et al. A novel bispecific antibody format enables simultaneous bivalent and monovalent co-engagement of distinct target antigens. MAbs 2011; 3:546-57; PMID: 22123055).

Other exemplary Fc mutations to promote heterodimerization of multispecific antibodies include those described in the following references, the contents of each of which is incorporated by reference herein, WO2016071377A1, US20140079689A1, US20160194389A1, US20160257763, WO2016071376A2, WO2015107026A1, WO2015107025A1, WO2015107015A1, US20150353636A1, US20140199294A1, U.S. Pat. No. 7,750,128B2, US20160229915A1, US20150344570A1, U.S. Pat. No. 8,003,774A1, US20150337049A1, US20150175707A1, US20140242075A1, US20130195849A1, US20120149876A1, US20140200331A1, U.S. Pat. No. 9,309,311B2, U.S. Pat. No. 8,586,713, US20140037621A1, US20130178605A1, US20140363426A1, US20140051835A1 and US20110054151A1.

Stabilizing cysteine mutations have also been used in combination with KiH and other Fc heterodimerization promoting variants, see e.g., U.S. Pat. No. 7,183,076. Other exemplary cysteine modifications include, e.g., those disclosed in US20140348839A1, U.S. Pat. No. 7,855,275B2, and U.S. Pat. No. 9,000,130B2.

Strand Exchange Engineered Domains (SEED)

Heterodimeric Fc platform that support the design of bispecific and asymmetric fusion proteins by devising strand-exchange engineered domain (SEED) C(H)3 heterodimers are known. These derivatives of human IgG and IgA C(H)3 domains create complementary human SEED C(H)3 heterodimers that are composed of alternating segments of human IgA and IgG C(H)3 sequences. The resulting pair of SEED C(H)3 domains preferentially associates to form heterodimers when expressed in mammalian cells. SEEDbody (Sb) fusion proteins consist of [IgG1 hinge]-C(H)2-[SEED C(H)3], that may be genetically linked to one or more fusion partners (see e.g., Davis J H et al. SEEDbodies: fusion proteins based on strand exchange engineered domain (SEED) CH3 heterodimers in an Fc analogue platform for asymmetric binders or immunofusions and bispecific antibodies. Protein Eng Des Sel 2010; 23:195-202; PMID:20299542 and U.S. Pat. No. 8,871,912. The contents of each of which are incorporated by reference herein).

Duobody

“Duobody” technology to produce bispecific antibodies with correct heavy chain pairing are known. The DuoBody technology involves three basic steps to generate stable bispecific human IgG1antibodies in a post-production exchange reaction. In a first step, two IgG1s, each containing single matched mutations in the third constant (CH3) domain, are produced separately using standard mammalian recombinant cell lines. Subsequently, these IgG1 antibodies are purified according to standard processes for recovery and purification. After production and purification (post-production), the two antibodies are recombined under tailored laboratory conditions resulting in a bispecific antibody product with a very high yield (typically >95%) (see e.g., Labrijn et al, PNAS 2013; 110(13):5145-5150 and Labrijn et al. Nature Protocols 2014; 9(10):2450-63, the contents of each of which are incorporated by reference herein).

Electrostatic Interactions

Methods of making multispecific antibodies using CH3 amino acid changes with charged amino acids such that homodimer formation is electrostatically unfavorable are disclosed. EP1870459 and WO 2009089004 describe other strategies for favoring heterodimer formation upon co-expression of different antibody domains in a host cell. In these methods, one or more residues that make up the heavy chain constant domain 3 (CH3), CH3-CH3 interfaces in both CH3 domains are replaced with a charged amino acid such that homodimer formation is electrostatically unfavorable and heterodimerization is electrostatically favorable. Additional methods of making multispecific molecules using electrostatic interactions are described in the following references, the contents of each of which is incorporated by reference herein, include US20100015133, U.S. Pat. No. 8,592,562B2, U.S. Pat. No. 9,200,060B2, US20140154254A1, and U.S. Pat. No. 9,358,286A1.

Common Light Chain

Light chain mispairing needs to be avoided to generate homogenous preparations of bispecific IgGs. One way to achieve this is through the use of the common light chain principle, i.e. combining two binders that share one light chain but still have separate specificities. An exemplary method of enhancing the formation of a desired bispecific antibody from a mixture of monomers is by providing a common variable light chain to interact with each of the heteromeric variable heavy chain regions of the bispecific antibody. Compositions and methods of producing bispecific antibodies with a common light chain as disclosed in, e.g., U.S. Pat. No. 7,183,076B2, US20110177073A1, EP2847231A1, WO2016079081A1, and EP3055329A1, the contents of each of which is incorporated by reference herein.

CrossMab

Another option to reduce light chain mispairing is the CrossMab technology which avoids non-specific L chain mispairing by exchanging CH1 and CL domains in the Fab of one half of the bispecific antibody. Such crossover variants retain binding specificity and affinity, but make the two arms so different that L chain mispairing is prevented. The CrossMab technology (as reviewed in Klein et al. Supra) involves domain swapping between heavy and light chains so as to promote the formation of the correct pairings. Briefly, to construct a bispecific IgG-like CrossMab antibody that could bind to two antigens by using two distinct light chain—heavy chain pairs, a two-step modification process is applied. First, a dimerization interface is engineered into the C-terminus of each heavy chain using a heterodimerization approach, e.g., Knob-into-hole (KiH) technology, to ensure that only a heterodimer of two distinct heavy chains from one antibody (e.g., Antibody A) and a second antibody (e.g., Antibody B) is efficiently formed. Next, the constant heavy 1 (CH1) and constant light (CL) domains of one antibody are exchanged (Antibody A), keeping the variable heavy (VH) and variable light (VL) domains consistent. The exchange of the CH1 and CL domains ensured that the modified antibody (Antibody A) light chain would only efficiently dimerize with the modified antibody (antibody A) heavy chain, while the unmodified antibody (Antibody B) light chain would only efficiently dimerize with the unmodified antibody (Antibody B) heavy chain; and thus only the desired bispecific CrossMab would be efficiently formed (see e.g., Cain, C. SciBX 4(28); doi:10.1038/scibx.2011.783, the contents of which are incorporated by reference herein).

Common Heavy Chain

An exemplary method of enhancing the formation of a desired bispecific antibody from a mixture of monomers is by providing a common variable heavy chain to interact with each of the heteromeric variable light chain regions of the bispecific antibody. Compositions and methods of producing bispecific antibodies with a common heavy chain are disclosed in, e.g., US20120184716, US20130317200, and US20160264685A1, the contents of each of which is incorporated by reference herein.

Amino Acid Modifications

Alternative compositions and methods of producing multispecific antibodies with correct light chain pairing include various amino acid modifications. For example, Zymeworks describes heterodimers with one or more amino acid modifications in the CH1 and/or CL domains, one or more amino acid modifications in the VH and/or VL domains, or a combination thereof, which are part of the interface between the light chain and heavy chain and create preferential pairing between each heavy chain and a desired light chain such that when the two heavy chains and two light chains of the heterodimer pair are co-expressed in a cell, the heavy chain of the first heterodimer preferentially pairs with one of the light chains rather than the other (see e.g., WO2015181805). Other exemplary methods are described in WO2016026943 (Argen-X), US20150211001, US20140072581A1, US20160039947A1, and US20150368352.

Lambda/Kappa Formats

Multispecific molecules (e.g., multispecific antibody molecules) that include the lambda light chain polypeptide and a kappa light chain polypeptides, can be used to allow for heterodimerization. Methods for generating bispecific antibody molecules comprising the lambda light chain polypeptide and a kappa light chain polypeptides are disclosed in PCT/US17/53053 filed on Sep. 22, 2017 and designated publication number WO 2018/057955, incorporated herein by reference in its entirety.

In embodiments, the multispecific molecule includes a multispecific antibody molecule, e.g., an antibody molecule comprising two binding specificities, e.g., a bispecific antibody molecule. The multispecific antibody molecule includes:

    • a lambda light chain polypeptide 1 (LLCP1) specific for a first epitope;
    • a heavy chain polypeptide 1 (HCP1) specific for the first epitope;
    • a kappa light chain polypeptide 2 (KLCP2) specific for a second epitope; and
    • a heavy chain polypeptide 2 (HCP2) specific for the second epitope.

“Lambda light chain polypeptide 1 (LLCP1)”, as that term is used herein, refers to a polypeptide comprising sufficient light chain (LC) sequence, such that when combined with a cognate heavy chain variable region, can mediate specific binding to its epitope and complex with an HCP1. In an embodiment it comprises all or a fragment of a CH1 region. In an embodiment, an LLCP1 comprises LC-CDR1, LC-CDR2, LC-CDR3, FR1, FR2, FR3, FR4, and CH1, or sufficient sequence therefrom to mediate specific binding of its epitope and complex with an HCP1. LLCP1, together with its HCP1, provide specificity for a first epitope (while KLCP2, together with its HCP2, provide specificity for a second epitope). As described elsewhere herein, LLCP1 has a higher affinity for HCP1 than for HCP2.

“Kappa light chain polypeptide 2 (KLCP2)”, as that term is used herein, refers to a polypeptide comprising sufficient light chain (LC) sequence, such that when combined with a cognate heavy chain variable region, can mediate specific binding to its epitope and complex with an HCP2. In some embodiments, it comprises all or a fragment of a CH1 region. In an embodiment, a KLCP2 comprises LC-CDR1, LC-CDR2, LC-CDR3, FR1, FR2, FR3, FR4, and CH1, or sufficient sequence therefrom to mediate specific binding of its epitope and complex with an HCP2. KLCP2, together with its HCP2, provide specificity for a second epitope (while LLCP1, together with its HCP1, provide specificity for a first epitope).

“Heavy chain polypeptide 1 (HCP1)”, as that term is used herein, refers to a polypeptide comprising sufficient heavy chain (HC) sequence, e.g., HC variable region sequence, such that when combined with a cognate LLCP1, can mediate specific binding to its epitope and complex with an HCP1. In some embodiments, it comprises all or a fragment of a CH1 region. In an embodiment, it comprises all or a fragment of a CH2 and/or CH3 region. In an embodiment an HCP1 comprises HC-CDR1, HC-CDR2, HC-CDR3, FR1, FR2, FR3, FR4, CH1, CH2, and CH3, or sufficient sequence therefrom to: (i) mediate specific binding of its epitope and complex with an LLCP1, (ii) to complex preferentially, as described herein to LLCP1 as opposed to KLCP2; and (iii) to complex preferentially, as described herein, to an HCP2, as opposed to another molecule of HCP1. HCP1, together with its LLCP1, provide specificity for a first epitope (while KLCP2, together with its HCP2, provide specificity for a second epitope).

“Heavy chain polypeptide 2 (HCP2)”, as that term is used herein, refers to a polypeptide comprising sufficient heavy chain (HC) sequence, e.g., HC variable region sequence, such that when combined with a cognate LLCP1, can mediate specific binding to its epitope and complex with an HCP1. In some embodiments, it comprises all or a fragment of a CH1region. In some embodiments, it comprises all or a fragment of a CH2 and/or CH3 region. In an embodiment an HCP1 comprises HC-CDR1, HC-CDR2, HC-CDR3, FR1, FR2, FR3, FR4, CH1, CH2, and CH3, or sufficient sequence therefrom to: (i) mediate specific binding of its epitope and complex with an KLCP2, (ii) to complex preferentially, as described herein to KLCP2 as opposed to LLCP1; and (iii) to complex preferentially, as described herein, to an HCP1, as opposed to another molecule of HCP2. HCP2, together with its KLCP2, provide specificity for a second epitope (while LLCP1, together with its HCP1, provide specificity for a first epitope).

In some embodiments of the multispecific antibody molecule disclosed herein:

    • LLCP1 has a higher affinity for HCP1 than for HCP2; and/or
    • KLCP2 has a higher affinity for HCP2 than for HCP1.

In embodiments, the affinity of LLCP1 for HCP1 is sufficiently greater than its affinity for HCP2, such that under preselected conditions, e.g., in aqueous buffer, e.g., at pH 7, in saline, e.g., at pH 7, or under physiological conditions, at least 75, 80, 90, 95, 98, 99, 99.5, or 99.9% of the multispecific antibody molecule molecules have a LLCP1 complexed, or interfaced with, a HCP1.

In some embodiments of the multispecific antibody molecule disclosed herein:

    • the HCP1 has a greater affinity for HCP2, than for a second molecule of HCP1; and/or
    • the HCP2 has a greater affinity for HCP1, than for a second molecule of HCP2.

In embodiments, the affinity of HCP1 for HCP2 is sufficiently greater than its affinity for a second molecule of HCP1, such that under preselected conditions, e.g., in aqueous buffer, e.g., at pH 7, in saline, e.g., at pH 7, or under physiological conditions, at least 75%, 80, 90, 95, 98, 99 99.5 or 99.9% of the multispecific antibody molecule molecules have a HCP1 complexed, or interfaced with, a HCP2.

In another aspect, disclosed herein is a method for making, or producing, a multispecific antibody molecule. The method includes:

    • (i) providing a first heavy chain polypeptide (e.g., a heavy chain polypeptide comprising one, two, three or all of a first heavy chain variable region (first VH), a first CH1, a first heavy chain constant region (e.g., a first CH2, a first CH3, or both));
    • (ii) providing a second heavy chain polypeptide (e.g., a heavy chain polypeptide comprising one, two, three or all of a second heavy chain variable region (second VH), a second CH1, a second heavy chain constant region (e.g., a second CH2, a second CH3, or both));
    • (iii) providing a lambda chain polypeptide (e.g., a lambda light variable region (VL□), a lambda light constant chain (VL□), or both) that preferentially associates with the first heavy chain polypeptide (e.g., the first VH); and
    • (iv) providing a kappa chain polypeptide (e.g., a lambda light variable region (VL□), a lambda light constant chain (VL□), or both) that preferentially associates with the second heavy chain polypeptide (e.g., the second VH), under conditions where (i)-(iv) associate.

In embodiments, the first and second heavy chain polypeptides form an Fc interface that enhances heterodimerization.

In embodiments, (i)-(iv) (e.g., nucleic acid encoding (i)-(iv)) are introduced in a single cell, e.g., a single mammalian cell, e.g., a CHO cell. In embodiments, (i)-(iv) are expressed in the cell.

In embodiments, (i)-(iv) (e.g., nucleic acid encoding (i)-(iv)) are introduced in different cells, e.g., different mammalian cells, e.g., two or more CHO cell. In embodiments, (i)-(iv) are expressed in the cells.

In embodiments, the method further comprises purifying a cell-expressed antibody molecule, e.g., using a lambda- and/or-kappa-specific purification, e.g., affinity chromatography.

In embodiments, the method further comprises evaluating the cell-expressed multispecific antibody molecule. For example, the purified cell-expressed multispecific antibody molecule can be analyzed by techniques known in the art, include mass spectrometry. In one embodiment, the purified cell-expressed antibody molecule is cleaved, e.g., digested with papain to yield the Fab moieties and evaluated using mass spectrometry.

In embodiments, the method produces correctly paired kappa/lambda multispecific, e.g., bispecific, antibody molecules in a high yield, e.g., at least 75%, 80, 90, 95, 98, 99 99.5 or 99.9%.

In other embodiments, the multispecific, e.g., a bispecific, antibody molecule that includes:

    • (i) a first heavy chain polypeptide (HCP1) (e.g., a heavy chain polypeptide comprising one, two, three or all of a first heavy chain variable region (first VH), a first CH1, a first heavy chain constant region (e.g., a first CH2, a first CH3, or both)), e.g., wherein the HCP1 binds to a first epitope;
    • (ii) a second heavy chain polypeptide (HCP2) (e.g., a heavy chain polypeptide comprising one, two, three or all of a second heavy chain variable region (second VH), a second CH1, a second heavy chain constant region (e.g., a second CH2, a second CH3, or both)), e.g., wherein the HCP2 binds to a second epitope;
    • (iii) a lambda light chain polypeptide (LLCP1) (e.g., a lambda light variable region (VL1), a lambda light constant chain (VL1), or both) that preferentially associates with the first heavy chain polypeptide (e.g., the first VH), e.g., wherein the LLCP1 binds to a first epitope; and (iv) a kappa light chain polypeptide (KLCP2) (e.g., a lambda light variable region (VLk), a lambda light constant chain (VLk), or both) that preferentially associates with the second heavy chain polypeptide (e.g., the second VH), e.g., wherein the KLCP2 binds to a second epitope.

In embodiments, the first and second heavy chain polypeptides form an Fc interface that enhances heterodimerization. In embodiments, the multispecific antibody molecule has a first binding specificity that includes a hybrid VL1-CL1 heterodimerized to a first heavy chain variable region connected to the Fc constant, CH2-CH3 domain (having a knob modification) and a second binding specificity that includes a hybrid VLk-CLk heterodimerized to a second heavy chain variable region connected to the Fc constant, CH2-CH3 domain (having a hole modification).

Cytokine Molecules

Cytokines are generally polypeptides that influence cellular activity, for example, through signal transduction pathways. Accordingly, a cytokine of the multispecific or multifunctional polypeptide is useful and can be associated with receptor-mediated signaling that transmits a signal from outside the cell membrane to modulate a response within the cell. Cytokines are proteinaceous signaling compounds that are mediators of the immune response. They control many different cellular functions including proliferation, differentiation and cell survival/apoptosis; cytokines are also involved in several pathophysiological processes including viral infections and autoimmune diseases. Cytokines are synthesized under various stimuli by a variety of cells of both the innate (monocytes, macrophages, dendritic cells) and adaptive (T- and B-cells) immune systems. Cytokines can be classified into two groups: pro- and anti-inflammatory. Pro-inflammatory cytokines, including IFNγ, IL-1, IL-6 and TNF-alpha, are predominantly derived from the innate immune cells and Th1 cells. Anti-inflammatory cytokines, including IL-10, IL-4, IL-13 and IL-5, are synthesized from Th2 immune cells.

The present disclosure provides, inter alia, multispecific (e.g., bi-, tri-, quad-specific) or multifunctional molecules, that include, e.g., are engineered to contain, one or more cytokine molecules, e.g., immunomodulatory (e.g., proinflammatory) cytokines and variants, e.g., functional variants, thereof. Accordingly, in some embodiments, the cytokine molecule is an interleukin or a variant, e.g., a functional variant thereof. In some embodiments the interleukin is a proinflammatory interleukin. In some embodiments the interleukin is chosen from interleukin-2 (IL-2), interleukin-12 (IL-12), interleukin-15 (IL-15), interleukin-18 (IL-18), interleukin-21 (IL-21), interleukin-7 (IL-7), or interferon gamma. In some embodiments, the cytokine molecule is a proinflammatory cytokine.

In certain embodiments, the cytokine is a single chain cytokine. In certain embodiments, the cytokine is a multichain cytokine (e.g., the cytokine comprises 2 or more (e.g., 2) polypeptide chains. An exemplary multichain cytokine is IL-12.

Examples of useful cytokines include, but are not limited to, GM-CSF, IL-1α, IL-1β, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-10, IL-12, IL-21, IFN-α, IFN-β, IFN-γ, MIP-1α, MIP-1β, TGF-β, TNF-α, and TNF-β. In one embodiment the cytokine of the multispecific or multifunctional polypeptide is a cytokine selected from the group of GM-CSF, IL-2, IL-7, IL-8, IL-10, IL-12, IL-15, IL-21, IFN-α, IFN-γ, MIP-1α, MIP-1β and TGF-β. In one embodiment the cytokine of the i the multispecific or multifunctional polypeptide is a cytokine selected from the group of IL-2, IL-7, IL-10, IL-12, IL-15, IFN-α, and IFN-γ. In certain embodiments the cytokine is mutated to remove N- and/or O-glycosylation sites. Elimination of glycosylation increases homogeneity of the product obtainable in recombinant production.

In one embodiment, the cytokine of the multispecific or multifunctional polypeptide is IL-2. In a specific embodiment, the IL-2 cytokine can elicit one or more of the cellular responses selected from the group consisting of: proliferation in an activated T lymphocyte cell, differentiation in an activated T lymphocyte cell, cytotoxic T cell (CTL) activity, proliferation in an activated B cell, differentiation in an activated B cell, proliferation in a natural killer (NK) cell, differentiation in a NK cell, cytokine secretion by an activated T cell or an NK cell, and NK/lymphocyte activated killer (LAK) antitumor cytotoxicity. In another particular embodiment the IL-2 cytokine is a mutant IL-2 cytokine having reduced binding affinity to the .alpha.-subunit of the IL-2 receptor. Together with the .beta.- and .gamma.-subunits (also known as CD122 and CD132, respectively), the .alpha.-subunit (also known as CD25) forms the heterotrimeric high-affinity IL-2 receptor, while the dimeric receptor consisting only of the β- and γ-subunits is termed the intermediate-affinity IL-2 receptor. As described in PCT patent application number PCT/EP2012/051991, which is incorporated herein by reference in its entirety, a mutant IL-2 polypeptide with reduced binding to the .alpha.-subunit of the IL-2 receptor has a reduced ability to induce IL-2 signaling in regulatory T cells, induces less activation-induced cell death (AICD) in T cells, and has a reduced toxicity profile in vivo, compared to a wild-type IL-2 polypeptide. The use of such an cytokine with reduced toxicity is particularly advantageous in a multispecific or multifunctional polypeptide according to the invention, having a long serum half-life due to the presence of an Fc domain. In one embodiment, the mutant IL-2 cytokine of the multispecific or multifunctional polypeptide according to the invention comprises at least one amino acid mutation that reduces or abolishes the affinity of the mutant IL-2 cytokine to the .alpha.-subunit of the IL-2 receptor (CD25) but preserves the affinity of the mutant IL-2 cytokine to the intermediate-affinity IL-2 receptor (consisting of the β and γ subunits of the IL-2 receptor), compared to the non-mutated IL-2 cytokine. In one embodiment the one or more amino acid mutations are amino acid substitutions. In a specific embodiment, the mutant IL-2 cytokine comprises one, two or three amino acid substitutions at one, two or three position(s) selected from the positions corresponding to residue 42, 45, and 72 of human IL-2. In a more specific embodiment, the mutant IL-2 cytokine comprises three amino acid substitutions at the positions corresponding to residue 42, 45 and 72 of human IL-2. In an even more specific embodiment, the mutant IL-2 cytokine is human IL-2 comprising the amino acid substitutions F42A, Y45A and L72G. In one embodiment the mutant IL-2 cytokine additionally comprises an amino acid mutation at a position corresponding to position 3 of human IL-2, which eliminates the O-glycosylation site of IL-2. Particularly, said additional amino acid mutation is an amino acid substitution replacing a threonine residue by an alanine residue. A particular mutant IL-2 cytokine useful in the invention comprises four amino acid substitutions at positions corresponding to residues 3, 42, 45 and 72 of human IL-2. Specific amino acid substitutions are T3A, F42A, Y45A and L72G. As demonstrated in PCT patent application number PCT/EP2012/051991 and in the appended Examples, said quadruple mutant IL-2 polypeptide (IL-2 qm) exhibits no detectable binding to CD25, reduced ability to induce apoptosis in T cells, reduced ability to induce IL-2 signaling in T.sub.reg cells, and a reduced toxicity profile in vivo. However, it retains ability to activate IL-2 signaling in effector cells, to induce proliferation of effector cells, and to generate IFN-γ as a secondary cytokine by NK cells.

The IL-2 or mutant IL-2 cytokine according to any of the above embodiments may comprise additional mutations that provide further advantages such as increased expression or stability. For example, the cysteine at position 125 may be replaced with a neutral amino acid such as alanine, to avoid the formation of disulfide-bridged IL-2 dimers. Thus, in certain embodiments the IL-2 or mutant IL-2 cytokine of the multispecific or multifunctional polypeptide according to the invention comprises an additional amino acid mutation at a position corresponding to residue 125 of human IL-2. In one embodiment said additional amino acid mutation is the amino acid substitution C125A.

In a specific embodiment the IL-2 cytokine of the multispecific or multifunctional polypeptide comprises the polypeptide sequence of SEQ ID NO: 2270

[APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKK ATELKHLQCLEEELKPLEEVLNLAQSKNFHLRPRDLISNINVIVLELKG SETTFMCEYADETATIVEFLNRWITFAQSIISTLT].

In another specific embodiment the IL-2 cytokine of the multispecific or multifunctional polypeptide comprises the polypeptide sequence of SEQ ID NO: 2280

[APASSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFAMPKKA TELKHLQCLEEELKPLEEVLNGAQSKNFHLRPRDLISNINVIVLELKGSE TTFMCEYADETATIVEFLNRWITFAQSIISTLT].

In another embodiment the cytokine of the multispecific or multifunctional polypeptide is IL-12. In a specific embodiment said IL-12 cytokine is a single chain IL-12 cytokine. In an even more specific embodiment the single chain IL-12 cytokine comprises the polypeptide sequence of SEQ ID NO: 2290 [IWELKKDVYVVELDWYPDAPGEMVVLTCDTPEEDGITWTLDQS SEVLGSGKTLTIQVKEF GDAGQYTCHKGGEVLSHSLLLLHKKEDGIWSTDILKDQKEPKNKTFLRCEAKNYSGRFTC WWLTTISTDLTFSVKSSRGSSDPQGVTCGAATLSAERVRGDNKEYEYSVECQEDSACPAAE ESLPIEVMVDAVHKLKYENYTSSFFIRDIIKPDPPKNLQLKPLKNSRQVEVSWEYPDTWSTP HSYFSLTFCVQVQGKSKREKKDRVFTDKTSATVICRKNASISVRAQDRYYSSSWSEWASVP CSGGGGSGGGGSGGGGSRNLPVATPDPGMFPCLHHSQNLLRAVSNMLQKARQTLEFYPCT SEEIDHEDITKDKTSTVEACLPLELTKNESCLNSRETSFITNGSCLASRKTSFMMALCLSSIYE DLKMYQVEFKTMNAKLLMDPKRQIFLDQNMLAVIDELMQALNFNSETVPQKSSLEEPDFY KTKIKLCILLHAFRIRAVTIDRVMSYLNAS]. In one embodiment, the IL-12 cytokine can elicit one or more of the cellular responses selected from the group consisting of: proliferation in a NK cell, differentiation in a NK cell, proliferation in a T cell, and differentiation in a T cell.

In another embodiment the cytokine of the multispecific or multifunctional polypeptide is IL-10. In a specific embodiment said IL-10 cytokine is a single chain IL-10 cytokine. In an even more specific embodiment the single chain IL-10 cytokine comprises the polypeptide sequence of SEQ ID NO: 2300

[SPGQGTQSENSCTHFPGNLPNMLRDLRDAFSRVKTFFQMKDQLDNLLLK ESLLEDFKGYLGCQALSEMIQFYLEEVMPQAENQDPDIKAHVNSLGENLK TLRLRLRRCHRFLPCENKSKAVEQVKNAFNKLQEKGIYKAMSEFDIFINY IEAYMTMKIRNGGGGSGGGGSGGGGSGGGGSSPGQGTQSENSCTHFPGNL PNMLRDLRDAFSRVKTFFQMKDQLDNLLLKESLLEDFKGYLGCQALSEMI QFYLEEVMPQAENQDPDIKAHVNSLGENLKTLRLRLRRCHRFLPCENKSK AVEQVKNAFNKLQEKGIYKAMSEFDIFINYIEAYMTMKIRN].

In another specific embodiment the IL-10 cytokine is a monomeric IL-10 cytokine. In a more specific embodiment the monomeric IL-10 cytokine comprises the polypeptide sequence of SEQ ID NO: 2310 [SPGQGTQSENSCTHFPGNLPNMLRDLRDAFSRVKTFFQMKDQLDNLLLKESLLEDFKGYL GCQALSEMIQFYLEEVMPQAENQDPDIKAHVNSLGENLKTLRLRLRRCHRFLPCENGGGSG GKSKAVEQVKNAFNKLQEKGIYKAMSEFDIFINYIEAYMTMKIRN]. In one embodiment, the IL-10 cytokine can elicit one or more of the cellular responses selected from the group consisting of: inhibition of cytokine secretion, inhibition of antigen presentation by antigen presenting cells, reduction of oxygen radical release, and inhibition of T cell proliferation. A multispecific or multifunctional polypeptide according to the invention wherein the cytokine is IL-10 is particularly useful for downregulation of inflammation, e.g. in the treatment of an inflammatory disorder.

In another embodiment, the cytokine of the multispecific or multifunctional polypeptide is IL-15. In a specific embodiment said IL-15 cytokine is a mutant IL-15 cytokine having reduced binding affinity to the α-subunit of the IL-15 receptor. Without wishing to be bound by theory, a mutant IL-15 polypeptide with reduced binding to the .alpha.-subunit of the IL-15 receptor has a reduced ability to bind to fibroblasts throughout the body, resulting in improved pharmacokinetics and toxicity profile, compared to a wild-type IL-15 polypeptide. The use of an cytokine with reduced toxicity, such as the described mutant IL-2 and mutant IL-15 effector moieties, is particularly advantageous in a multispecific or multifunctional polypeptide according to the invention, having a long serum half-life due to the presence of an Fc domain. In one embodiment the mutant IL-15 cytokine of the multispecific or multifunctional polypeptide according to the invention comprises at least one amino acid mutation that reduces or abolishes the affinity of the mutant IL-15 cytokine to the .alpha.-subunit of the IL-15 receptor but preserves the affinity of the mutant IL-15 cytokine to the intermediate-affinity IL-15/IL-2 receptor (consisting of the .beta.- and .gamma.-subunits of the IL-15/IL-2 receptor), compared to the non-mutated IL-15 cytokine. In one embodiment the amino acid mutation is an amino acid substitution. In a specific embodiment, the mutant IL-15 cytokine comprises an amino acid substitution at the position corresponding to residue 53 of human IL-15. In a more specific embodiment, the mutant IL-15 cytokine is human IL-15 comprising the amino acid substitution E53A. In one embodiment the mutant IL-15 cytokine additionally comprises an amino acid mutation at a position corresponding to position 79 of human IL-15, which eliminates the N-glycosylation site of IL-15. Particularly, said additional amino acid mutation is an amino acid substitution replacing an asparagine residue by an alanine residue. In an even more specific embodiment the IL-15 cytokine comprises the polypeptide sequence of SEQ ID NO: 2320 [NWVNVISDLKKIEDLIQSMHIDATLYTESDVHPSCKVTAMKCFLLELQVISLASGDASIHDT VENLIILANNSLSSNGAVTESGCKECEELEEKNIKEFLQSFVHIVQMFINTS]. In one embodiment, the IL-15 cytokine can elicit one or more of the cellular responses selected from the group consisting of: proliferation in an activated T lymphocyte cell, differentiation in an activated T lymphocyte cell, cytotoxic T cell (CTL) activity, proliferation in an activated B cell, differentiation in an activated B cell, proliferation in a natural killer (NK) cell, differentiation in a NK cell, cytokine secretion by an activated T cell or an NK cell, and NK/lymphocyte activated killer (LAK) antitumor cytotoxicity.

Mutant cytokine molecules useful as effector moieties in the multispecific or multifunctional polypeptide can be prepared by deletion, substitution, insertion or modification using genetic or chemical methods well known in the art. Genetic methods may include site-specific mutagenesis of the encoding DNA sequence, PCR, gene synthesis, and the like. The correct nucleotide changes can be verified for example by sequencing. Substitution or insertion may involve natural as well as non-natural amino acid residues. Amino acid modification includes well known methods of chemical modification such as the addition or removal of glycosylation sites or carbohydrate attachments, and the like.

In one embodiment, the cytokine, particularly a single-chain cytokine, of the multispecific or multifunctional polypeptide is GM-CSF. In a specific embodiment, the GM-CSF cytokine can elicit proliferation and/or differentiation in a granulocyte, a monocyte or a dendritic cell. In one embodiment, the cytokine, particularly a single-chain cytokine, of the multispecific or multifunctional polypeptide is IFN-α. In a specific embodiment, the IFN-α cytokine can elicit one or more of the cellular responses selected from the group consisting of: inhibiting viral replication in a virus-infected cell, and upregulating the expression of major histocompatibility complex I (MHC I). In another specific embodiment, the IFN-α cytokine can inhibit proliferation in a tumor cell. In one embodiment the cytokine, particularly a single-chain cytokine, of the multispecific or multifunctional polypeptide is IFNγ. In a specific embodiment, the IFN-γ cytokine can elicit one or more of the cellular responses selected from the group of: increased macrophage activity, increased expression of MHC molecules, and increased NK cell activity. In one embodiment the cytokine, particularly a single-chain cytokine, of the multispecific or multifunctional polypeptide is IL-7. In a specific embodiment, the IL-7 cytokine can elicit proliferation of T and/or B lymphocytes. In one embodiment, the cytokine, particularly a single-chain cytokine, of the multispecific or multifunctional polypeptide is IL-8. In a specific embodiment, the IL-8 cytokine can elicit chemotaxis in neutrophils. In one embodiment, the cytokine, particularly a single-chain cytokine, of the multispecific or multifunctional polypeptide, is MIP-1α. In a specific embodiment, the MIP-1αcytokine can elicit chemotaxis in monocytes and T lymphocyte cells. In one embodiment, the cytokine, particularly a single-chain cytokine, of the multispecific or multifunctional polypeptide is MIP-1β. In a specific embodiment, the MIP-1β cytokine can elicit chemotaxis in monocytes and T lymphocyte cells. In one embodiment, the cytokine, particularly a single-chain cytokine, of the multispecific or multifunctional polypeptide is TGF-β. In a specific embodiment, the TGF-β cytokine can elicit one or more of the cellular responses selected from the group consisting of: chemotaxis in monocytes, chemotaxis in macrophages, upregulation of IL-1 expression in activated macrophages, and upregulation of IgA expression in activated B cells.

In one embodiment, the multispecific or multifunctional polypeptide of the invention binds to an cytokine receptor with a dissociation constant (KD) that is at least about 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5 or 10 times greater than that for a control cytokine. In another embodiment, the multispecific or multifunctional polypeptide binds to an cytokine receptor with a KD that is at least 2, 3, 4, 5, 6, 7, 8, 9, or 10 times greater than that for a corresponding multispecific or multifunctional polypeptide comprising two or more effector moieties. In another embodiment, the multispecific or multifunctional polypeptide binds to an cytokine receptor with a dissociation constant KD that is about 10 times greater than that for a corresponding the multispecific or multifunctional polypeptide comprising two or more cytokines.

In some embodiments, the multispecific molecules disclosed herein include a cytokine molecule. In embodiments, the cytokine molecule includes a full length, a fragment or a variant of a cytokine; a cytokine receptor domain, e.g., a cytokine receptor dimerizing domain; or an agonist of a cytokine receptor, e.g., an antibody molecule (e.g., an agonistic antibody) to a cytokine receptor.

In some embodiments the cytokine molecule is chosen from IL-2, IL-12, IL-15, IL-18, IL-7, IL-21, or interferon gamma, or a fragment or variant thereof, or a combination of any of the aforesaid cytokines. The cytokine molecule can be a monomer or a dimer. In embodiments, the cytokine molecule can further include a cytokine receptor dimerizing domain.

In other embodiments, the cytokine molecule is an agonist of a cytokine receptor, e.g., an antibody molecule (e.g., an agonistic antibody) to a cytokine receptor chosen from an IL-15Ra or IL-21R.

In one embodiment, the cytokine molecule is IL-15, e.g., human IL-15 (e.g., comprising the amino acid sequence: NWVNVISDLKKIEDLIQSMHIDATLYTESDVHPSCKVTAMKCFLLELQVISLESGDASIHDT VENLIILANNSLSSNGNVTESGCKECEELEEKNIKEFLQSFVHIVQMFINTS (SEQ ID NO: 2170), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 2170.

In some embodiments, the cytokine molecule comprises a receptor dimerizing domain, e.g., an IL15Ralpha dimerizing domain. In one embodiment, the IL15Ralpha dimerizing domain comprises the amino acid sequence: MAPRRARGCRTLGLPALLLLLLLRPPATRGITCPPPMSVEHADIWVKSYSLYSRERYICNSG FKRKAGTSSLTECVL (SEQ ID NO: 2180), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 2180. In some embodiments, the cytokine molecule (e.g., IL-15) and the receptor dimerizing domain (e.g., an IL15Ralpha dimerizing domain) of the multispecific molecule are covalently linked, e.g., via a linker (e.g., a Gly-Ser linker, e.g., a linker comprising the amino acid sequence SGGSGGGGSGGGSGGGGSLQ (SEQ ID NO: 2190). In other embodiments, the cytokine molecule (e.g., IL-15) and the receptor dimerizing domain (e.g., an IL15Ralpha dimerizing domain) of the multispecific molecule are not covalently linked, e.g., are non-covalently associated.

In other embodiments, the cytokine molecule is IL-2, e.g., human IL-2 (e.g., comprising the amino acid sequence: APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLEE ELKPLEEVLNLAQSKNFHLRPRDLISNINVIVLELKGSETTFMCEYADETATIVEFLNRWITF CQSIISTLT (SEQ ID NO: 2191), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO:2191).

In other embodiments, the cytokine molecule is IL-18, e.g., human IL-18 (e.g., comprising the amino acid sequence: YFGKLESKLSVIRNLNDQVLFIDQGNRPLFEDMTDSDCRDNAPRTIFIISMYKDSQPRGMAV TISVKCEKISTLSCENKIISFKEMNPPDNIKDTKSDIIFFQRSVPGHDNKMQFESSSYEGYFLA CEKERDLFKLILKKEDELGDRSIMFTVQNED (SEQ ID NO: 2192), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 2192).

In other embodiments, the cytokine molecule is IL-21, e.g., human IL-21 (e.g., comprising the amino acid sequence: QGQDRHMIRMRQLIDIVDQLKNYVNDLVPEFLPAPEDVETNCEWSAFSCFQKAQLKSANT GNNERIINVSIKKLKRKPPSTNAGRRQKHRLTCPSCDSYEKKPPKEFLERFKSLLQKMIHQH LSSRTHGSEDS (SEQ ID NO: 2193), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 2193).

In yet other embodiments, the cytokine molecule is interferon gamma, e.g., human interferon gamma (e.g., comprising the amino acid sequence: QDPYVKEAENLKKYFNAGHSDVADNGTLFLGILKNWKEESDRKMQSQIVSFYFKLFKNF KDDQSIQKSVETIKEDMNVKFFNSNKKKRDDFEKLTNYSVTDLNVQRKAIHELIQVMAELS PAAKTGKRKRSQMLFRG (SEQ ID NO: 2194), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 2194).

Immune Cell Engagers

The immune cell engagers, e.g., first and/or second immune cell engager, of the multispecific or multifunctional molecules disclosed herein can mediate binding to, and/or activation of, an immune cell, e.g., an immune effector cell. In some embodiments, the immune cell is chosen from a T cell, an NK cell, a B cell, a dendritic cell, or a macrophage cell engager, or a combination thereof. In some embodiments, the immune cell engager is chosen from one, two, three, or all of a T cell engager, NK cell engager, a B cell engager, a dendritic cell engager, or a macrophage cell engager, or a combination thereof. The immune cell engager can be an agonist of the immune system. In some embodiments, the immune cell engager can be an antibody molecule, a ligand molecule (e.g., a ligand that further comprises an immunoglobulin constant region, e.g., an Fc region), a small molecule, a nucleotide molecule.

Natural Killer Cell Engagers

Natural Killer (NK) cells recognize and destroy tumors and virus-infected cells in an antibody-independent manner. The regulation of NK cells is mediated by activating and inhibiting receptors on the NK cell surface. One family of activating receptors is the natural cytotoxicity receptors (NCRs) which include NKp30, NKp44 and NKp46. The NCRs initiate tumor targeting by recognition of heparan sulfate on cancer cells. NKG2D is a receptor that provides both stimulatory and costimulatory innate immune responses on activated killer (NK) cells, leading to cytotoxic activity. DNAM1 is a receptor involved in intercellular adhesion, lymphocyte signaling, cytotoxicity and lymphokine secretion mediated by cytotoxic T-lymphocyte (CTL) and NK cell. DAP10 (also known as HCST) is a transmembrane adapter protein which associates with KLRK1 to form an activation receptor KLRK1-HCST in lymphoid and myeloid cells; this receptor plays a major role in triggering cytotoxicity against target cells expressing cell surface ligands such as WIC class I chain-related MICA and MICB, and U (optionally L1)6-binding proteins (ULBPs); it KLRK1-HCST receptor plays a role in immune surveillance against tumors and is required for cytolysis of tumors cells; indeed, melanoma cells that do not express KLRK1 ligands escape from immune surveillance mediated by NK cells. CD16 is a receptor for the Fc region of IgG, which binds complexed or aggregated IgG and also monomeric IgG and thereby mediates antibody-dependent cellular cytotoxicity (ADCC) and other antibody-dependent responses, such as phagocytosis.

In some embodiments, the NK cell engager is a viral hemagglutinin (HA), HA is a glycoprotein found on the surface of influenza viruses. It is responsible for binding the virus to cells with sialic acid on the membranes, such as cells in the upper respiratory tract or erythrocytes. HA has at least 18 different antigens. These subtypes are named H1 through H18. NCRs can recognize viral proteins. NKp46 has been shown to be able to interact with the HA of influenza and the HA-NA of Paramyxovirus, including Sendai virus and Newcastle disease virus. Besides NKp46, NKp44 can also functionally interact with HA of different influenza subtypes.

The present disclosure provides, inter alia, multispecific (e.g., bi-, tri-, quad-specific) or multifunctional molecules, that are engineered to contain one or more NK cell engagers that mediate binding to and/or activation of an NK cell. Accordingly, in some embodiments, the NK cell engager is selected from an antigen binding domain or ligand that binds to (e.g., activates): NKp30, NKp40, NKp44, NKp46, NKG2D, DNAM1, DAP10, CD16 (e.g., CD16a, CD16b, or both), CRTAM, CD27, PSGL1, CD96, CD100 (SEMA4D), NKp80, CD244 (also known as SLAMF4 or 2B4), SLAMF6, SLAMF7, KIR2DS2, KIR2DS4, KIR3DS1, KIR2DS3, KIR2DS5, KIR2DS1, CD94, NKG2C, NKG2E, or CD160.

In one embodiment, the NK cell engager is a ligand of NKp30 is a B7-6, e.g., comprises the amino acid sequence of:

DLKVEMMAGGTQITPLNDNVTIFCNIFYSQPLNITSMGITWFWKSLTFDKEVKVF EFFGDHQEAFRPGAIVSPWRLKSGDASLRLPGIQLEEAGEYRCEVVVTPLKAQGTVQLEVV ASPASRLLLDQVGMKENEDKYMCESSGFYPEAINITWEKQTQKFPHPIEISEDVITGPTIKN MDGTFNVTSCLKLNSSQEDPGTVYQCVVRHASLHTPLRSNFTLTAARHSLSETEKTDNFS (SEQ ID NO: 3291), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3291.

In other embodiments, the NK cell engager is a ligand of NKp44 or NKp46, which is a viral HA. Viral hemagglutinins (HA) are glyco proteins which are on the surface of viruses. HA proteins allow viruses to bind to the membrane of cells via sialic acid sugar moieties which contributes to the fusion of viral membranes with the cell membranes (see e.g., Eur J Immunol. 2001 September; 31(9):2680-9 “Recognition of viral hemagglutinins by NKp44 but not by NKp30”; and Nature. 2001 Feb. 22; 409(6823):1055-60 “Recognition of haemagglutinins on virus-infected cells by NKp46 activates lysis by human NK cells” the contents of each of which are incorporated by reference herein).

In other embodiments, the NK cell engager is a ligand of NKG2D chosen from MICA, MICB, or ULBP1, e.g., wherein:

    • (i) MICA comprises the amino acid sequence:

(SEQ ID NO: 3292) EPHSLRYNLTVLSWDGSVQSGFLTEVHLDGQPFLRCDRQKCRAKPQGQWA EDVLGNKTWDRETRDLTGNGKDLRMTLAHIKDQKEGLHSLQEIRVCEIHE DNSTRSSQHFYYDGELFLSQNLETKEWTMPQSSRAQTLAMNVRNFLKEDA MKTKTHYHAMHADCLQELRRYLKSGVVLRRTVPPMVNVTRSEASEGNITV TCRASGFYPWNITLSWRQDGVSLSHDTQQWGDVLPDGNGTYQTWVATRIC QGEEQRFTCYMEHSGNHSTHPVPSGKVLVLQSHW,
    • a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3292;
    • (ii) MICB comprises the amino acid sequence:

(SEQ ID NO: 3293) AEPHSLRYNLMVLSQDESVQSGFLAEGHLDGQPFLRYDRQKRRAKPQGQW AEDVLGAKTWDTETEDLTENGQDLRRTLTHIKDQKGGLHSLQEIRVCEIH EDSSTRGSRHFYYDGELFLSQNLETQESTVPQSSRAQTLAMNVTNFWKED AMKTKTHYRAMQADCLQKLQRYLKSGVAIRRTVPPMVNVTCSEVSEGNIT VTCRASSFYPRNITLTWRQDGVSLSHNTQQWGDVLPDGNGTYQTWVATRI RQGEEQRFTCYMEHSGNHGTHPVPSGKVLVLQSQRTD,
    • a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3293; or
    • (iii) ULBP1 comprises the amino acid sequence: GWVDTHCLCYDFIITPKSRPEPQWCEVQGLVDERPFLHYDCVNHKAKAFASLGKKVNVTK TWEEQTETLRDVVDFLKGQLLDIQVENLIPIEPLTLQARMSCEHEAHGHGRGSWQFLFNGQ KFLLFDSNNRKWTALHPGAKKMTEKWEKNRDVTMFFQKISLGDCKMWLEEFLMYWEQM LDPTKPPSLAPG (SEQ ID NO: 3294), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3294.

In other embodiments, the NK cell engager is a ligand of DNAM1 chosen from NECTIN2 or NECL5, e.g., wherein:

    • (i) NECTIN2 comprises the amino acid sequence: QDVRVQVLPEVRGQLGGTVELPCHLLPPVPGLYISLVTWQRPDAPANHQNVAAFHPKMGP SFPSPKPGSERLSFVSAKQSTGQDTEAELQDATLALHGLTVEDEGNYTCEFATFPKGSVRG MTWLRVIAKPKNQAEAQKVTFSQDPTTVALCISKEGRPPARISWLSSLDWEAKETQVSGTL AGTVTVTSRFTLVPSGRADGVTVTCKVEHESFEEPALIPVTLSVRYPPEVSISGYDDNWYLG RTDATLSCDVRSNPEPTGYDWSTTSGTFPTSAVAQGSQLVIHAVDSLFNTTFVCTVTNAVG MGRAEQVIFVRETPNTAGAGATGG (SEQ ID NO: 3295), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3295; or
    • (ii) NECL5 comprises the amino acid sequence: WPPPGTGDVVVQAPTQVPGFLGDSVTLPCYLQVPNMEVTHVSQLTWARHGESGSMAVFH QTQGPSYSESKRLEFVAARLGAELRNASLRMFGLRVEDEGNYTCLFVTFPQGSRSVDIWLR VLAKPQNTAEVQKVQLTGEPVPMARCVSTGGRPPAQITWHSDLGGMPNTSQVPGFLSGTV TVTSLWILVPSSQVDGKNVTCKVEHESFEKPQLLTVNLTVYYPPEVSISGYDNNWYLGQNE ATLTCDARSNPEPTGYNWSTTMGPLPPFAVAQGAQLLIRPVDKPINTTLICNVTNALGARQ AELTVQVKEGPPSEHSGISRN (SEQ ID NO: 3296), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3296.

In yet other embodiments, the NK cell engager is a ligand of DAP10, which is an adapter for NKG2D (see e.g., Proc Natl Acad Sci USA. 2005 May 24; 102(21): 7641-7646; and Blood, 15 Sep. 2011 Volume 118, Number 11, the full contents of each of which is incorporated by reference herein).

In other embodiments, the NK cell engager is a ligand of CD16, which is a CD16a/b ligand, e.g., a CD16a/b ligand further comprising an antibody Fc region (see e.g., Front Immunol. 2013; 4: 76 discusses how antibodies use the Fc to trigger NK cells through CD16, the full contents of which are incorporated herein).

In other embodiments, the NK cell engager is a ligand of CRTAM, which is NECL2, e.g., wherein NECL2 comprises the amino acid sequence: QNLFTKDVTVIEGEVATISCQVNKSDDSVIQLLNPNRQTIYFRDFRPLKDSRFQLLNFSSSEL KVSLTNVSISDEGRYFCQLYTDPPQESYTTITVLVPPRNLMIDIQKDTAVEGEEIEVNCTAM ASKPATTIRWFKGNTELKGKSEVEEW SDMYTVTSQLMLKVHKEDDGVPVICQVEHPAVT GNLQTQRYLEVQYKPQVHIQMTYPLQGLTREGDALELTCEAIGKPQPVMVTWVRVDDEM PQHAVLSGPNLFINNLNKTDNGTYRCEASNIVGKAHSDYMLYVYDPPTTIPPPTTTTTTTTT TTTTILTIITDSRAGEEGSIRAVDH (SEQ ID NO: 3297), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3297.

In other embodiments, the NK cell engager is a ligand of CD27, which is CD70, e.g., wherein CD70 comprises the amino acid sequence: QRFAQAQQQLPLESLGWDVAELQLNHTGPQQDPRLYWQGGPALGRSFLHGPELDKGQLRI HRDGIYMVHIQVTLAICSSTTASRHHPTTLAVGICSPASRSISLLRLSFHQGCTIASQRLTPLA RGDTLCTNLTGTLLPSRNTDETFFGVQWVRP (SEQ ID NO: 3298), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3298.

In other embodiments, the NK cell engager is a ligand of PSGL1, which is L-selectin (CD62L), e.g., wherein L-selectin comprises the amino acid sequence:

(SEQ ID NO: 3299) WTYHYSEKPMNWQRARRFCRDNYTDLVAIQNKAEIEYLEKTLPFSRSYYW IGIRKIGGIWTWVGTNKSLTEEAENWGDGEPNNKKNKEDCVEIYIKRNKD AGKWNDDACHKLKAALCYTASCQPWSCSGHGECVEIINNYTCNCDVGYYG PQCQFVIQCEPLEAPELGTMDCTHPLGNFSFSSQCAFSCSEGTNLTGIEE TTCGPFGNWSSPEPTCQVIQCEPLSAPDLGIMNCSHPLASFSFTSACTFI CSEGTELIGKKKTICESSGIWSNPSPICQKLDKSFSMIKEGDYN,

a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3299.

In other embodiments, the NK cell engager is a ligand of CD96, which is NECL5, e.g., wherein NECL5 comprises the amino acid sequence: WPPPGTGDVVVQAPTQVPGFLGDSVTLPCYLQVPNMEVTHVSQLTWARHGESGSMAVFH QTQGPSYSESKRLEFVAARLGAELRNASLRMFGLRVEDEGNYTCLFVTFPQGSRSVDIWLR VLAKPQNTAEVQKVQLTGEPVPMARCVSTGGRPPAQITWHSDLGGMPNTSQVPGFLSGTV TVTSLWILVPSSQVDGKNVTCKVEHESFEKPQLLTVNLTVYYPPEVSISGYDNNWYLGQNE ATLTCDARSNPEPTGYNWSTTMGPLPPFAVAQGAQLLIRPVDKPINTTLICNVTNALGARQ AELTVQVKEGPPSEHSGISRN (SEQ ID NO: 3296), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3296.

In other embodiments, the NK cell engager is a ligand of CD100 (SEMA4D), which is CD72, e.g., wherein CD72 comprises the amino acid sequence: RYLQVSQQLQQTNRVLEVTNSSLRQQLRLKITQLGQSAEDLQGSRRELAQSQEALQVEQR AHQAAEGQLQACQADRQKTKETLQSEEQQRRALEQKLSNMENRLKPFFTCGSADTCCPSG WIMHQKSCFYISLTSKNWQESQKQCETLSSKLATF SEIYPQ SHSYYFLNSLLPNGGSGNSYW TGLSSNKDWKLTDDTQRTRTYAQSSKCNKVHKTWSWWTLESESCRSSLPYICEMTAFRFP D (SEQ ID NO: 3300), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3300.

In other embodiments, the NK cell engager is a ligand of NKp80, which is CLEC2B (AICL), e.g., wherein CLEC2B (AICL) comprises the amino acid sequence: KLTRDSQSLCPYDWIGFQNKCYYFSKEEGDWNSSKYNCSTQHADLTIIDNIEEM NFLRRYKCSSDHWIGLKMAKNRTGQWVDGATFTKSFGMRGSEGCAYLSDDGAATARCY TERKWICRKRIH (SEQ ID NO: 3301), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3301.

In other embodiments, the NK cell engager is a ligand of CD244, which is CD48, e.g., wherein CD48 comprises the amino acid sequence: QGHLVHMTVVSGSNVTLNISESLPENYKQLTWFYTFDQKIVEWDSRKSKYFESKFKGRVR LDPQSGALYISKVQKEDNSTYIMRVLKKTGNEQEWKIKLQVLDPVPKPVIKIEKIEDMDDN CYLKLSCVIPGESVNYTWYGDKRPFPKELQNSVLETTLMPHNYSRCYTCQVSNSVSSKNGT VCLSPPCTLARS (SEQ ID NO: 3302), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3302.

T Cell Engagers

The present disclosure provides, inter alia, multispecific (e.g., bi-, tri-, quad-specific) or multifunctional molecules, that are engineered to contain one or more T cell engager that mediate binding to and/or activation of a T cell. In some embodiments, the T cell engager is an antigen binding domain that binds to, e.g., activates TCRβ, e.g., a TCRβV region, as described herein. In some embodiments, the T cell engager is selected from an antigen binding domain or ligand that binds to (e.g., and in some embodiments activates) one or more of CD3, TCRα, TCRy, TCR, ICOS, CD28, CD27, HVEM, LIGHT, CD40, 4-1BB, OX40, DR3, GITR, CD30, TIM1, SLAM, CD2, or CD226. In other embodiments, the T cell engager is selected from an antigen binding domain or ligand that binds to and does not activate one or more of CD3, TCRα, TCRγ, TCRζ, ICOS, CD28, CD27, HVEM, LIGHT, CD40, 4-1BB, OX40, DR3, GITR, CD30, TIM1, SLAM, CD2, or CD226.

B Cell, Macrophage & Dendritic Cell Engagers

Broadly, B cells, also known as B lymphocytes, are a type of white blood cell of the lymphocyte subtype. They function in the humoral immunity component of the adaptive immune system by secreting antibodies. Additionally, B cells present antigen (they are also classified as professional antigen-presenting cells (APCs)) and secrete cytokines. Macrophages are a type of white blood cell that engulfs and digests cellular debris, foreign substances, microbes, cancer cells via phagocytosis. Besides phagocytosis, they play important roles in nonspecific defense (innate immunity) and also help initiate specific defense mechanisms (adaptive immunity) by recruiting other immune cells such as lymphocytes. For example, they are important as antigen presenters to T cells. Beyond increasing inflammation and stimulating the immune system, macrophages also play an important anti-inflammatory role and can decrease immune reactions through the release of cytokines. Dendritic cells (DCs) are antigen-presenting cells that function in processing antigen material and present it on the cell surface to the T cells of the immune system.

The present disclosure provides, inter alia, multispecific (e.g., bi-, tri-, quad-specific) or multifunctional molecules, that include, e.g., are engineered to contain, one or more B cell, macrophage, and/or dendritic cell engager that mediate binding to and/or activation of a B cell, macrophage, and/or dendritic cell.

Accordingly, in some embodiments, the immune cell engager comprises a B cell, macrophage, and/or dendritic cell engager chosen from one or more of CD40 ligand (CD40L) or a CD70 ligand; an antibody molecule that binds to CD40 or CD70; an antibody molecule to OX40; an OX40 ligand (OX40L); an agonist of a Toll-like receptor (e.g., as described herein, e.g., a TLR4, e.g., a constitutively active TLR4 (caTLR4), or a TLR9 agonists); a 41BB; a CD2; a CD47; or a STING agonist, or a combination thereof.

In some embodiments, the B cell engager is a CD40L, an OX40L, or a CD70 ligand, or an antibody molecule that binds to OX40, CD40 or CD70.

In some embodiments, the macrophage engager is a CD2 agonist. In some embodiments, the macrophage engager is an antigen binding domain that binds to: CD40L or antigen binding domain or ligand that binds CD40, a Toll like receptor (TLR) agonist (e.g., as described herein), e.g., a TLR9 or TLR4 (e.g., caTLR4 (constitutively active TLR4), CD47, or a STING agonist. In some embodiments, the STING agonist is a cyclic dinucleotide, e.g., cyclic di-GMP (cdGMP) or cyclic di-AMP (cdAMP). In some embodiments, the STING agonist is biotinylated.

In some embodiments, the dendritic cell engager is a CD2 agonist. In some embodiments, the dendritic cell engager is a ligand, a receptor agonist, or an antibody molecule that binds to one or more of: OX40L, 41BB, a TLR agonist (e.g., as described herein) (e.g., TLR9 agonist, TLR4 (e.g., caTLR4 (constitutively active TLR4)), CD47, or and a STING agonist. In some embodiments, the STING agonist is a cyclic dinucleotide, e.g., cyclic di-GMP (cdGMP) or cyclic di-AMP (cdAMP). In some embodiments, the STING agonist is biotinylated.

In other embodiments, the immune cell engager mediates binding to, or activation of, one or more of a B cell, a macrophage, and/or a dendritic cell. Exemplary B cell, macrophage, and/or dendritic cell engagers can be chosen from one or more of CD40 ligand (CD40L) or a CD70 ligand; an antibody molecule that binds to CD40 or CD70; an antibody molecule to OX40; an OX40 ligand (OX40L); a Toll-like receptor agonist (e.g., a TLR4, e.g., a constitutively active TLR4 (caTLR4) or a TLR9 agonist); a 41BB agonist; a CD2; a CD47; or a STING agonist, or a combination thereof.

In some embodiments, the B cell engager is chosen from one or more of a CD40L, an OX40L, or a CD70 ligand, or an antibody molecule that binds to OX40, CD40 or CD70.

In other embodiments, the macrophage cell engager is chosen from one or more of a CD2 agonist; a CD40L; an OX40L; an antibody molecule that binds to OX40, CD40 or CD70; a Toll-like receptor agonist or a fragment thereof (e.g., a TLR4, e.g., a constitutively active TLR4 (caTLR4)); a CD47 agonist; or a STING agonist.

In other embodiments, the dendritic cell engager is chosen from one or more of a CD2 agonist, an OX40 antibody, an OX40L, 41BB agonist, a Toll-like receptor agonist or a fragment thereof (e.g., a TLR4, e.g., a constitutively active TLR4 (caTLR4)), CD47 agonist, or a STING agonist.

In one embodiment, the OX40L comprises the amino acid sequence: QVSHRYPRIQSIKVQFTEYKKEKGFILTSQKEDEIMKVQNNSVIINCDGFYLISLKGYFSQEV NISLHYQKDEEPLFQLKKVRSVNSLMVASLTYKDKVYLNVTTDNTSLDDFHVNGGELILIH QNPGEFCVL (SEQ ID NO: 3303), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3303.

In another embodiment, the CD40L comprises the amino acid sequence: MQKGDQNPQIAAHVISEASSKTTSVLQWAEKGYYTMSNNLVTLENGKQLTVKRQGLYYI YAQVTFCSNREASSQAPFIASLCLKSPGRFERILLRAANTHSSAKPCGQQSIHLGGVFELQPG ASVFVNVTDPSQVSHGTGFTSFGLLKL (SEQ ID NO: 3304), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3304.

In yet other embodiments, the STING agonist comprises a cyclic dinucleotide, e.g., a cyclic di-GMP (cdGMP), a cyclic di-AMP (cdAMP), or a combination thereof, optionally with 2′,5′ or 3′,5′ phosphate linkages.

In one embodiment, the immune cell engager includes 41BB ligand, e.g., comprising the amino acid sequence: ACPWAVSGARASPGSAASPRLREGPELSPDDPAGLLDLRQGMFAQLVAQNVLLIDGPLSW YSDPGLAGVSLTGGLSYKEDTKELVVAKAGVYYVFFQLELRRVVAGEGSGSVSLALHLQP LRSAAGAAALALTVDLPPASSEARNSAFGFQGRLLHLSAGQRLGVHLHTEARARHAWQLT QGATVLGLFRVTPEIPAGLPSPRSE (SEQ ID NO: 3305), a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3305.

Toll-Like Receptors

Toll-Like Receptors (TLRs) are evolutionarily conserved receptors are homologues of the Drosophila Toll protein, and recognize highly conserved structural motifs known as pathogen-associated microbial patterns (PAMPs), which are exclusively expressed by microbial pathogens, or danger-associated molecular patterns (DAMPs) that are endogenous molecules released from necrotic or dying cells. PAMPs include various bacterial cell wall components such as lipopolysaccharide (LPS), peptidoglycan (PGN) and lipopeptides, as well as flagellin, bacterial DNA and viral double-stranded RNA. DAMPs include intracellular proteins such as heat shock proteins as well as protein fragments from the extracellular matrix. Stimulation of TLRs by the corresponding PAMPs or DAMPs initiates signaling cascades leading to the activation of transcription factors, such as AP-1, NF-κB and interferon regulatory factors (IRFs). Signaling by TLRs results in a variety of cellular responses, including the production of interferons (IFNs), pro-inflammatory cytokines and effector cytokines that direct the adaptive immune response. TLRs are implicated in a number of inflammatory and immune disorders and play a role in cancer (Rakoff-Nahoum S. & Medzhitov R., 2009. Toll-like receptors and cancer. Nat Revs Cancer 9:57-63.)

TLRs are type I transmembrane proteins characterized by an extracellular domain containing leucine-rich repeats (LRRs) and a cytoplasmic tail that contains a conserved region called the Toll/IL-1 receptor (TIR) domain. Ten human and twelve murine TLRs have been characterized, TLR1 to TLR10 in humans, and TLR1 to TLR9, TLR11, TLR12 and TLR13 in mice, the homolog of TLR10 being a pseudogene. TLR2 is essential for the recognition of a variety of PAMPs from Gram-positive bacteria, including bacterial lipoproteins, lipomannans and lipoteichoic acids. TLR3 is implicated in virus-derived double-stranded RNA. TLR4 is predominantly activated by lipopolysaccharide. TLR5 detects bacterial flagellin and TLR9 is required for response to unmethylated CpG DNA. Finally, TLR7 and TLR8 recognize small synthetic antiviral molecules, and single-stranded RNA was reported to be their natural ligand. TLR11 has been reported to recognize uropathogenic E. coli and a profilin-like protein from Toxoplasma gondii. The repertoire of specificities of the TLRs is apparently extended by the ability of TLRs to heterodimerize with one another. For example, dimers of TLR2 and TLR6 are required for responses to diacylated lipoproteins while TLR2 and TLR1 interact to recognize triacylated lipoproteins. Specificities of the TLRs are also influenced by various adapter and accessory molecules, such as MD-2 and CD14 that form a complex with TLR4 in response to LPS.

TLR signaling consists of at least two distinct pathways: a MyD88-dependent pathway that leads to the production of inflammatory cytokines, and a MyD88-independent pathway associated with the stimulation of IFN-β and the maturation of dendritic cells. The MyD88-dependent pathway is common to all TLRs, except TLR3 (Adachi O. et al., 1998. Targeted disruption of the MyD88 gene results in loss of IL-1- and IL-18-mediated function. Immunity. 9(1):143-50). Upon activation by PAMPs or DAMPs, TLRs hetero- or homodimerize inducing the recruitment of adaptor proteins via the cytoplasmic TIR domain. Individual TLRs induce different signaling responses by usage of the different adaptor molecules. TLR4 and TLR2 signaling requires the adaptor TIRAP/Mal, which is involved in the MyD88-dependent pathway. TLR3 triggers the production of IFN-β in response to double-stranded RNA, in a MyD88-independent manner, through the adaptor TRIF/TICAM-1. TRAM/TICAM-2 is another adaptor molecule involved in the MyD88-independent pathway which function is restricted to the TLR4 pathway.

TLR3, TLR7, TLR8 and TLR9 recognize viral nucleic acids and induce type I IFNs. The signaling mechanisms leading to the induction of type I IFNs differ depending on the TLR activated. They involve the interferon regulatory factors, IRFs, a family of transcription factors known to play a critical role in antiviral defense, cell growth and immune regulation. Three IRFs (IRF3, IRF5 and IRF7) function as direct transducers of virus-mediated TLR signaling. TLR3 and TLR4 activate IRF3 and IRF7, while TLR7 and TLR8 activate IRF5 and IRF7 (Doyle S. et al., 2002. IRF3 mediates a TLR3/TLR4-specific antiviral gene program. Immunity. 17(3):251-63). Furthermore, type I IFN production stimulated by TLR9 ligand CpG-A has been shown to be mediated by PI(3)K and mTOR (Costa-Mattioli M. & Sonenberg N. 2008. RAPping production of type I interferon in pDCs through mTOR. Nature Immunol. 9: 1097-1099).

TLR-9

TLR9 recognizes unmethylated CpG sequences in DNA molecules. CpG sites are relatively rare (˜1%) on vertebrate genomes in comparison to bacterial genomes or viral DNA. TLR9 is expressed by numerous cells of the immune system such as B lymphocytes, monocytes, natural killer (NK) cells, and plasmacytoid dendritic cells. TLR9 is expressed intracellularly, within the endosomal compartments and functions to alert the immune system of viral and bacterial infections by binding to DNA rich in CpG motifs. TLR9 signals leads to activation of the cells initiating pro-inflammatory reactions that result in the production of cytokines such as type-I interferon and IL-12.

TLR Agonists

A TLR agonist can agonize one or more TLR, e.g., one or more of human TLR-1, 2, 3, 4, 5, 6, 7, 8, 9, or 10. In some embodiments, an adjunctive agent described herein is a TLR agonist. In some embodiments, the TLR agonist specifically agonizes human TLR-9. In some embodiments, the TLR-9 agonist is a CpG moiety. As used herein, a CpG moiety, is a linear dinucleotide having the sequence: 5′-C-phosphate-G-3′, that is, cytosine and guanine separated by only one phosphate.

In some embodiments, the CpG moiety comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, or more CpG dinucleotides. In some embodiments, the CpG moiety consists of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 CpG dinucleotides. In some embodiments, the CpG moiety has 1-5, 1-10, 1-20, 1-30, 1-40, 1-50, 5-10, 5-20, 5-30, 10-20, 10-30, 10-40, or 10-50 CpG dinucleotides.

In some embodiments, the TLR-9 agonist is a synthetic ODN (oligodeoxynucleotides). CpG ODNs are short synthetic single-stranded DNA molecules containing unmethylated CpG dinucleotides in particular sequence contexts (CpG motifs). CpG ODNs possess a partially or completely phosphorothioated (PS) backbone, as opposed to the natural phosphodiester (PO) backbone found in genomic bacterial DNA. There are three major classes of CpG ODNs: classes A, B and C, which differ in their immunostimulatory activities. CpG-A ODNs are characterized by a PO central CpG-containing palindromic motif and a PS-modified 3′ poly-G string. They induce high IFN-α production from pDCs but are weak stimulators of TLR9-dependent NF-κB signaling and pro-inflammatory cytokine (e.g. IL-6) production. CpG-B ODNs contain a full PS backbone with one or more CpG dinucleotides. They strongly activate B cells and TLR9-dependent NF-κB signaling but weakly stimulate IFN-α secretion. CpG-C ODNs combine features of both classes A and B. They contain a complete PS backbone and a CpG-containing palindromic motif. C-Class CpG ODNs induce strong IFN-α production from pDC as well as B cell stimulation.

Tumor-Targeting Moieties

The present disclosure provides, inter alia, multispecific (e.g., bi-, tri-, tetra-specific) molecules, that include, e.g., are engineered to contain, one or more tumor specific targeting moieties that direct the molecule to a tumor cell.

In certain embodiments, the multispecific molecules disclosed herein include a tumor-targeting moiety. The tumor targeting moiety can be chosen from an antibody molecule (e.g., an antigen binding domain as described herein), a receptor or a receptor fragment, or a ligand or a ligand fragment, or a combination thereof. In some embodiments, the tumor targeting moiety associates with, e.g., binds to, a tumor cell (e.g., a molecule, e.g., antigen, present on the surface of the tumor cell). In certain embodiments, the tumor targeting moiety targets, e.g., directs the multispecific molecules disclosed herein to a cancer (e.g., a cancer or tumor cells). In some embodiments, the cancer is chosen from a hematological cancer, a solid cancer, a metastatic cancer, or a combination thereof.

In some embodiments, the multispecific molecule, e.g., the tumor-targeting moiety, binds to a solid tumor antigen or a stromal antigen. The solid tumor antigen or stromal antigen can be present on a solid tumor, or a metastatic lesion thereof. In some embodiments, the solid tumor is chosen from one or more of pancreatic (e.g., pancreatic adenocarcinoma), breast, colorectal, lung (e.g., small or non-small cell lung cancer), skin, ovarian, or liver cancer. In one embodiment, the solid tumor is a fibrotic or desmoplastic solid tumor. For example, the solid tumor antigen or stromal antigen can be present on a tumor, e.g., a tumor of a class typified by having one or more of: limited tumor perfusion, compressed blood vessels, or fibrotic tumor interstitium.

In certain embodiments, the solid tumor antigen is chosen from one or more of: PDL1, CD47, gangloside 2 (GD2), prostate stem cell antigen (PSCA), prostate specific membrane antigen (PMSA), prostate-specific antigen (PSA), carcinoembryonic antigen (CEA), Ron Kinase, c-Met, Immature laminin receptor, TAG-72, BING-4, Calcium-activated chloride channel 2, Cyclin-B1, 9D7, Ep-CAM, EphA3, Her2/neu, Telomerase, SAP-1, Survivin, NY-ESO-1/LAGE-1, PRAME, SSX-2, Melan-A/MART-1, Gp100/pme117, Tyrosinase, TRP-1/-2, MC1R, β-catenin, BRCA1/2, CDK4, CML66, Fibronectin, p53, Ras, TGF-B receptor, AFP, ETA, MAGE, MUC-1, CA-125, BAGE, GAGE, NY-ESO-1, β-catenin, CDK4, CDC27, CD47, α actinin-4, TRP1/gp75, TRP2, gp100, Melan-A/MART1, gangliosides, WT1, EphA3, Epidermal growth factor receptor (EGFR), MART-2, MART-1, MUC1, MUC2, MUM1, MUM2, MUM3, NA88-1, NPM, OA1, OGT, RCC, RUI1, RUI2, SAGE, TRG, TRP1, TSTA, Folate receptor alpha, L1-CAM, CAIX, EGFRvIII, gpA33, GD3, GM2, VEGFR, Intergrins (Integrin alphaVbeta3, Integrin alpha5Beta1), Carbohydrates (Le), IGF1R, EPHA3, TRAILR1, TRAILR2, or RANKL.

In other embodiments, the multispecific molecule, e.g., the tumor-targeting moiety, binds to a molecule, e.g., antigen, present on the surface of a hematological cancer, e.g., a leukemia or a lymphoma. In some embodiments, the hematological cancer is a B-cell or T cell malignancy. In some embodiments, the hematological cancer is chosen from one or more of a Hodgkin's lymphoma, Non-Hodgkin's lymphoma (e.g., B cell lymphoma, diffuse large B cell lymphoma, follicular lymphoma, chronic lymphocytic leukemia, mantle cell lymphoma, marginal zone B-cell lymphoma, Burkitt lymphoma, lymphoplasmacytic lymphoma, hairy cell leukemia), acute myeloid leukemia (AML), chronic myeloid leukemia, myelodysplastic syndrome (MDS), multiple myeloma, or acute lymphocytic leukemia. In embodiments, the cancer is other than acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS). In embodiments, the hematological antigen is chosen from CD47, CD99, CD30, CD38, SLAMF7, or NY-ESO1. In some embodiments, the hematological antigen is chosen from is chosen from one or more of: BCMA, CD19, CD20, CD22, CD33, CD123, FcRH5, CLEC12, or CD179A.

Stromal Modifying Moieties

Solid tumors have a distinct structure that mimics that of normal tissues and comprises two distinct but interdependent compartments: the parenchyma (neoplastic cells) and the stroma that the neoplastic cells induce and in which they are dispersed. All tumors have stroma and require stroma for nutritional support and for the removal of waste products. In the case of tumors which grow as cell suspensions (e.g., leukemias, ascites tumors), the blood plasma serves as stroma (Connolly J L et al. Tumor Structure and Tumor Stroma Generation. In: Kufe D W et al., editors. Holland-Frei Cancer Medicine. 6th edition. Hamilton: BC Decker; 2003). The stroma includes a variety of cell types, including fibroblasts/myofibroblasts, glial, epithelial, fat, vascular, smooth muscle, and immune cells along with extracellular matrix (ECM) and extracellular molecules (Li Hanchen et al. Tumor Microenvironment: The Role of the Tumor Stroma in Cancer. J of Cellular Biochemistry 101: 805-815 (2007)).

Stromal modifying moieties described herein include moieties (e.g., proteins, e.g., enzymes) capable of degrading a component of the stroma, e.g., an ECM component, e.g., a glycosaminoglycan, e.g., hyaluronan (also known as hyaluronic acid or HA), chondroitin sulfate, chondroitin, dermatan sulfate, heparin sulfate, heparin, entactin, tenascin, aggrecan and keratin sulfate; or an extracellular protein, e.g., collagen, laminin, elastin, fibrinogen, fibronectin, and vitronectin.

Stromal Modifying Enzymes

In some embodiments, the stromal modifying moiety is an enzyme. For example, the stromal modifying moiety can include, but is not limited to a hyaluronidase, a collagenase, a chondroitinase, a matrix metalloproteinase (e.g., macrophage metalloelastase).

Hyaluronidases

Hyaluronidases are a group of neutral- and acid-active enzymes found throughout the animal kingdom. Hyaluronidases vary with respect to substrate specificity, and mechanism of action. There are three general classes of hyaluronidases: (1) Mammalian-type hyaluronidases, (EC 3.2.1.35) which are endo-beta-N-acetylhexosaminidases with tetrasaccharides and hexasaccharides as the major end products. They have both hydrolytic and transglycosidase activities, and can degrade hyaluronan and chondroitin sulfates; (2) Bacterial hyaluronidases (EC 4.2.99.1) degrade hyaluronan and, and to various extents, chondroitin sulfate and dermatan sulfate. They are endo-beta-N-acetylhexosaminidases that operate by a beta elimination reaction that yields primarily disaccharide end products; (3) Hyaluronidases (EC 3.2.1.36) from leeches, other parasites, and crustaceans are endo-beta-glucuronidases that generate tetrasaccharide and hexasaccharide end products through hydrolysis of the beta 1-3 linkage.

Mammalian hyaluronidases can be further divided into two groups: (1) neutral active and (2) acid active enzymes. There are six hyaluronidase-like genes in the human genome, HYAL1, HYAL2, HYAL3 HYAL4 HYALP1 and PH20/SPAM1. HYALP1 is a pseudogene, and HYAL3 has not been shown to possess enzyme activity toward any known substrates. HYAL4 is a chondroitinase and lacks activity towards hyaluronan. HYAL1 is the prototypical acid-active enzyme and PH20 is the prototypical neutral-active enzyme. Acid active hyaluronidases, such as HYAL1 and HYAL2 lack catalytic activity at neutral pH. For example, HYAL1 has no catalytic activity in vitro over pH 4.5 (Frost and Stern, “A Microtiter-Based Assay for Hyaluronidase Activity Not Requiring Specialized Reagents”, Analytical Biochemistry, vol. 251, pp. 263-269 (1997). HYAL2 is an acid active enzyme with a very low specific activity in vitro.

In some embodiments the hyaluronidase is a mammalian hyaluronidase. In some embodiments the hyaluronidase is a recombinant human hyaluronidase. In some embodiments, the hyaluronidase is a neutral active hyaluronidase. In some embodiments, the hyaluronidase is a neutral active soluble hyaluronidase. In some embodiments, the hyaluronidase is a recombinant PH20 neutral-active enzyme. In some embodiments, the hyaluronidase is a recombinant PH20 neutral-active soluble enzyme. In some embodiments the hyaluronidase is glycosylated. In some embodiments, the hyaluronidase possesses at least one N-linked glycan. A recombinant hyaluronidase can be produced using conventional methods known to those of skill in the art, e.g., U.S. Pat. No. 7,767,429, the entire contents of which are incorporated by reference herein.

In some embodiments the hyaluronidase is rHuPH20 (also referred to as Hylenex®; presently manufactured by Halozyme; approved by the FDA in 2005 (see e.g., Scodeller P (2014) Hyaluronidase and other Extracellular Matrix Degrading Enzymes for Cancer Therapy: New Uses and Nano-Formulations. J Carcinog Mutage 5:178; U.S. Pat. Nos. 7,767,429; 8,202,517; 7,431,380; 8,450,470; 8,772,246; 8,580,252, the entire contents of each of which is incorporated by reference herein). rHuPH20 is produced by genetically engineered CHO cells containing a DNA plasmid encoding for a soluble fragment of human hyaluronidase PH20. In some embodiments the hyaluronidase is glycosylated. In some embodiments, the hyaluronidase possesses at least one N-linked glycan. A recombinant hyaluronidase can be produced using conventional methods known to those of skill in the art, e.g., U.S. Pat. No. 7,767,429, the entire contents of which are incorporated by reference herein. In some embodiments, rHuPH20 has a sequence at least 95% (e.g., at least 96%, 97%, 98%, 99%, 100%) identical to the amino acid sequence of

(SEQ ID NO: 3306) LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSLFSFIGSPRINATG QGVTIFYVDRLGYYPYIDSITGVTVNGGIPQKISLQDHLDKAKKDITFYM PVDNLGMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNVQLSLTEAT EKAKQEFEKAGKDFLVETIKLGKLLRPNHLWGYYLFPDCYNHHYKKPGYN GSCFNVEIKRNDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRVREA IRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLSQDELVYTFGETVALGASG IVIWGTLSIMRSMKSCLLLDNYMETILNPYIINVTLAAKMCSQVLCQEQG VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPTLEDLEQFSEKFYC SCYSTLSCKEKADVKDTDAVDVCIADGVCIDAFLKPPMETEEPQIFYNAS PSTLS.

In any of the methods provided herein, the anti-hyaluronan agent can be an agent that degrades hyaluronan or can be an agent that inhibits the synthesis of hyaluronan. For example, the anti-hyaluronan agent can be a hyaluronan degrading enzyme. In another example, the anti-hyaluronan agent or is an agent that inhibits hyaluronan synthesis. For example, the anti-hyaluronan agent is an agent that inhibits hyaluronan synthesis such as a sense or antisense nucleic acid molecule against an HA synthase or is a small molecule drug. For example, an anti-hyaluronan agent is 4-methylumbelliferone (MU) or a derivative thereof, or leflunomide or a derivative thereof. Such derivatives include, for example, a derivative of 4-methylumbelliferone (MU) that is 6,7-dihydroxy-4-methyl coumarin or 5,7-dihydroxy-4-methyl coumarin.

In further examples of the methods provided herein, the hyaluronan degrading enzyme is a hyaluronidase. In some examples, the hyaluronan-degrading enzyme is a PH20 hyaluronidase or truncated form thereof to lacking a C-terminal glycosylphosphatidylinositol (GPI) attachment site or a portion of the GPI attachment site. In specific examples, the hyaluronidase is a PH20 selected from a human, monkey, bovine, ovine, rat, mouse or guinea pig PH20. For example, the hyaluronan-degrading enzyme is a human PH20 hyaluronidase that is neutral active and N-glycosylated and is selected from among (a) a hyaluronidase polypeptide that is a full-length PH20 or is a C-terminal truncated form of the PH20, wherein the truncated form includes at least amino acid residues 36-464 of SEQ ID NO: 139, such as 36-481, 36-482, 36-483, where the full-length PH20 has the sequence of amino acids set forth in SEQ ID NO: 139; or (b) a hyaluronidase polypeptide comprising a sequence of amino acids having at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity with the polypeptide or truncated form of sequence of amino acids set forth in SEQ ID NO: 139; or (c) a hyaluronidase polypeptide of (a) or (b) comprising amino acid substitutions, whereby the hyaluronidase polypeptide has a sequence of amino acids having at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity with the polypeptide set forth in SEQ ID NO: 139 or the with the corresponding truncated forms thereof. In exemplary examples, the hyaluronan-degrading enzyme is a PH20 that comprises a composition designated rHuPH20.

In other examples, the anti-hyaluronan agent is a hyaluronan degrading enzyme that is modified by conjugation to a polymer. The polymer can be a PEG and the anti-hyaluronan agent a PEGylated hyaluronan degrading enzyme. Hence, in some examples of the methods provided herein the hyaluronan-degrading enzyme is modified by conjugation to a polymer. For example, the hyaluronan-degrading enzyme is conjugated to a PEG, thus the hyaluronan degrading enzyme is PEGylated. In an exemplary example, the hyaluronan-degrading enzyme is a PEGylated PH20 enzyme (PEGPH20). In the methods provided herein, the corticosteroid can be a glucocorticoid that is selected from among cortisones, dexamethasones, hydrocortisones, methylprednisolones, prednisolones and prednisones.

Chondroitinases

Chondroitinases are enzymes found throughout the animal kingdom which degrade glycosaminoglycans, specifically chondroitins and chondroitin sulfates, through an endoglycosidase reaction. In some embodiments the chondroitinase is a mammalian chondroitinase. In some embodiments the chondroitinase is a recombinant human chondroitinase. In some embodiments the chondroitinase is HYAL4. Other exemplary chondroitinases include chondroitinase ABC (derived from Proteus vulgaris; Japanese Patent Application Laid-open No 6-153947, T. Yamagata et al. J. Biol. Chem., 243, 1523 (1968), S. Suzuki et al, J. Biol. Chem., 243, 1543 (1968)), chondroitinase AC (derived from Flavobacterium heparinum; T. Yamagata et al., J. Biol. Chem., 243, 1523 (1968)), chondroitinase AC II (derived from Arthrobacter aurescens; K. Hiyama, and S. Okada, J. Biol. Chem., 250, 1824 (1975), K. Hiyama and S. Okada, J. Biochem. (Tokyo), 80, 1201 (1976)), Hyaluronidase ACIII (derived from Flavobacterium sp. Hp102; Hirofumi Miyazono et al., Seikagaku, 61, 1023 (1989)), chondroitinase B (derived from Flavobacterium heparinum; Y. M. Michelacci and C. P. Dietrich, Biochem. Biophys. Res. Commun., 56, 973 (1974), Y. M. Michelacci and C. P. Dietrich, Biochem. J., 151, 121 (1975), Kenichi Maeyama et al, Seikagaku, 57, 1189 (1985)), chondroitinase C (derived from Flavobacterium sp. Hp102; Hirofumi Miyazono et al, Seikagaku, 61, 1023 (1939)), and the like.

Matrix Metalloproteinases

Matrix metalloproteases (MMPs) are zinc-dependent endopeptidases that are the major proteases involved in extracellular matrix (ECM) degradation. MMPs are capable of degrading a wide range of extracellular molecules and a number of bioactive molecules. Twenty-four MMP genes have been identified in humans, which can be organized into six groups based on domain organization and substrate preference: Collagenases (MMP-1, -8 and -13), Gelatinases (MMP-2 and MMP-9), Stromelysins (MMP-3, -10 and -11), Matrilysin (MMP-7 and MMP-26), Membrane-type (MT)-MMPs (MMP-14, -15, -16, -17, -24 and -25) and others (MMP-12, -19, -20, -21, -23, -27 and -28). In some embodiments, the stromal modifying moiety is a human recombinant MMP (e.g., MMP-1, -2, -3, -4, -5, -6, -7, -8, -9, 10, -11, -12, -13, -14, 15, -15, -17, -18, -19, 20, -21, -22, -23, or -24).

Collagenases

The three mammalian collagenases (MMP-1, -8, and -13) are the principal secreted endopeptidases capable of cleaving collagenous extracellular matrix. In addition to fibrillar collagens, collagenases can cleave several other matrix and non-matrix proteins including growth factors. Collagenases are synthesized as inactive pro-forms, and once activated, their activity is inhibited by specific tissue inhibitors of metalloproteinases, TIMPs, as well as by non-specific proteinase inhibitors (Ala-aho R et al. Biochimie. Collagenases in cancer. 2005 March-April; 87(3-4):273-86). In some embodiments, the stromal modifying moiety is a collagenase. In some embodiments, the collagenase is a human recombinant collagenase. In some embodiments, the collagenase is MMP-1. In some embodiments, the collagenase is MMP-8. In some embodiments, the collagenase is MMP-13.

Macrophage Metalloelastase

Macrophage metalloelastase (MME), also known as MMP-12, is a member of the stromelysin subgroup of MMPs and catalyzes the hydrolysis of soluble and insoluble elastin and a broad selection of matrix and nonmatrix substrates including type IV collagen, fibronectin, laminin, vitronectin, entactin, heparan, and chondroitin sulfates (Erja Kerkelä et al. Journal of Investigative Dermatology (2000) 114, 1113-1119; doi:10.1046/j.1523-1747.2000.00993). In some embodiments, the stromal modifying moiety is a MME. In some embodiments, the MME is a human recombinant MME. In some embodiments, the MME is MMP-12.

Additional Stromal Modifying Moieties

In some embodiments, the stromal modifying moiety causes one or more of: decreases the level or production of a stromal or extracellular matrix (ECM) component; decreases tumor fibrosis; increases interstitial tumor transport; improves tumor perfusion; expands the tumor microvasculature; decreases interstitial fluid pressure (IFP) in a tumor; or decreases or enhances penetration or diffusion of an agent, e.g., a cancer therapeutic or a cellular therapy, into a tumor or tumor vasculature.

In some embodiments, the stromal or ECM component decreased is chosen from a glycosaminoglycan or an extracellular protein, or a combination thereof. In some embodiments, the glycosaminoglycan is chosen from hyaluronan (also known as hyaluronic acid or HA), chondroitin sulfate, chondroitin, dermatan sulfate, heparin, heparin sulfate, entactin, tenascin, aggrecan and keratin sulfate. In some embodiments, the extracellular protein is chosen from collagen, laminin, elastin, fibrinogen, fibronectin, or vitronectin. In some embodiments, the stromal modifying moiety includes an enzyme molecule that degrades a tumor stroma or extracellular matrix (ECM). In some embodiments, the enzyme molecule is chosen from a hyaluronidase molecule, a collagenase molecule, a chondroitinase molecule, a matrix metalloproteinase molecule (e.g., macrophage metalloelastase), or a variant (e.g., a fragment) of any of the aforesaid. The term “enzyme molecule” includes a full length, a fragment or a variant of the enzyme, e.g., an enzyme variant that retains at least one functional property of the naturally-occurring enzyme.

In some embodiments, the stromal modifying moiety decreases the level or production of hyaluronic acid. In other embodiments, the stromal modifying moiety comprises a hyaluronan degrading enzyme, an agent that inhibits hyaluronan synthesis, or an antibody molecule against hyaluronic acid.

In some embodiments, the hyaluronan degrading enzyme is a hyaluronidase molecule, e.g., a full length or a variant (e.g., fragment thereof) thereof. In some embodiments, the hyaluronan degrading enzyme is active in neutral or acidic pH, e.g., pH of about 4-5. In some embodiments, the hyaluronidase molecule is a mammalian hyaluronidase molecule, e.g., a recombinant human hyaluronidase molecule, e.g., a full length or a variant (e.g., fragment thereof, e.g., a truncated form) thereof. In some embodiments, the hyaluronidase molecule is chosen from HYAL1, HYAL2, or PH-20/SPAM1, or a variant thereof (e.g., a truncated form thereof). In some embodiments, the truncated form lacks a C-terminal glycosylphosphatidylinositol (GPI) attachment site or a portion of the GPI attachment site. In some embodiments, the hyaluronidase molecule is glycosylated, e.g., comprises at least one N-linked glycan.

In some embodiments, the hyaluronidase molecule comprises the amino acid sequence: LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSLF SFIGSPRINATGQGVTIFYVDRLG YYPYIDSITGVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGMAVIDWEEWRPTWARN WKPKDVYKNRSIELVQQQNVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRPNHLWG YYLFPDCYNHHYKKPGYNGSCFNVEIKRNDDLSWLWNESTALYP SIYLNTQQ SPVAATLY VRNRVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLSQDELVYTFGETVALGASGIVIW GTLSIMRSMKSCLLLDNYMETILNPYIINVTLAAKMCSQVLCQEQGVCIRKNWNSSDYLHL NPDNFAIQLEKGGKFTVRGKPTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDVCIAD GVCIDAFLKPPMETEEPQIFYNASPSTLS (SEQ ID NO:3311), or a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3311.

In some embodiments, the hyaluronidase molecule comprises:

    • (i) the amino acid sequence of 36-464 of SEQ ID NO: 3311;
    • (ii) the amino acid sequence of 36-481, 36-482, or 36-483 of PH20, wherein PH20 has the sequence of amino acids set forth in SEQ ID NO: 3311; or
    • (iii) an amino acid sequence having at least 95% to 100% sequence identity to the polypeptide or truncated form of sequence of amino acids set forth in SEQ ID NO: 3311; or
    • (iv) an amino acid sequence having 30, 20, 10, 5 or fewer amino acid substitutions to the amino acid sequence set forth in SEQ ID NO: 3311. In some embodiments, the hyaluronidase molecule comprises an amino acid sequence at least 95% (e.g., at least 95%, 96%, 97%, 98%, 99%, 100%) identical to the amino acid sequence of SEQ ID NO: 3311. In some embodiments, the hyaluronidase molecule is encoded by a nucleotide sequence at least 95% (e.g., at least 96%, 97%, 98%, 99%, 100%) identical to the nucleotide sequence of SEQ ID NO: 3311.

In some embodiments, the hyaluronidase molecule is PH20, e.g., rHuPH20. In some embodiments, the hyaluronidase molecule is HYAL1 and comprises the amino acid sequence: FRGPLLPNRPFTTVWNANTQWCLERHGVDVDVSVFDVVANPGQTFRGPDMTIFYSSQGTY PYYTPTGEPVFGGLPQNASLIAHLARTFQDILAAIPAPDF SGLAVIDWEAWRPRWAFNWDT KDIYRQRSRALVQAQHPDWPAPQVEAVAQDQFQGAARAWMAGTLQLGRALRPRGLWGF YGFPDCYNYDFLSPNYTGQCPSGIRAQNDQLGWLWGQSRALYPSIYMPAVLEGTGKSQM YVQHRVAEAFRVAVAAGDPNLPVLPYVQIFYDTTNHFLPLDELEHSLGESAAQGAAGVVL WVSWENTRTKESCQAIKEYMDTTLGPFILNVTSGALLCSQALCSGHGRCVRRTSHPKALLL LNPASFSIQLTPGGGPLSLRGALSLEDQAQMAVEFKCRCYPGWQAPWCERKSMW (SEQ ID NO: 3312), or a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3312.

In some embodiments, the hyaluronan degrading enzyme, e.g., the hyaluronidase molecule, further comprises a polymer, e.g., is conjugated to a polymer, e.g., PEG. In some embodiments, the hyaluronan-degrading enzyme is a PEGylated PH20 enzyme (PEGPH20). In some embodiments, the hyaluronan degrading enzyme, e.g., the hyaluronidase molecule, further comprises an immunoglobulin chain constant region (e.g., Fc region) chosen from, e.g., the heavy chain constant regions of IgG1, IgG2, IgG3, and IgG4, more particularly, the heavy chain constant region of human IgG1, IgG2, IgG3, or IgG4. In some embodiments, the immunoglobulin constant region (e.g., the Fc region) is linked, e.g., covalently linked to, the hyaluronan degrading enzyme, e.g., the hyaluronidase molecule. In some embodiments, the immunoglobulin chain constant region (e.g., Fc region) is altered, e.g., mutated, to increase or decrease one or more of: Fc receptor binding, antibody glycosylation, the number of cysteine residues, effector cell function, or complement function. In some embodiments, the hyaluronan degrading enzyme, e.g., the hyaluronidase molecule forms a dimer.

In some embodiments, the stromal modifying moiety comprises an inhibitor of the synthesis of hyaluronan, e.g., an HA synthase. In some embodiments, the inhibitor comprises a sense or an antisense nucleic acid molecule against an HA synthase or is a small molecule drug. In some embodiments, the inhibitor is 4-methylumbelliferone (MU) or a derivative thereof (e.g., 6,7-dihydroxy-4-methyl coumarin or 5,7-dihydroxy-4-methyl coumarin), or leflunomide or a derivative thereof.

In some embodiments, the stromal modifying moiety comprises antibody molecule against hyaluronic acid.

In some embodiments, the stromal modifying moiety comprises a collagenase molecule, e.g., a mammalian collagenase molecule, or a variant (e.g., fragment) thereof. In some embodiments, the collagenase molecule is collagenase molecule IV, e.g., comprising the amino acid sequence of: YNFFPRKPKWDKNQITYRIIGYTPDLDPETVDDAFARAFQVWSDVTPLRFSRIHDGEADIMI NFGRWEHGDGYPFDGKDGLLAHAFAPGTGVGGDSHFDDDELWTLGEGQVVRVKYGNAD GEYCKFPFLFNGKEYNSCTDTGRSDGFLWCSTTYNFEKDGKYGFCPHEALFTMGGNAEGQ PCKFPFRFQGTSYDSCTTEGRTDGYRWCGTTEDYDRDKKYGFCPETAMSTVGGNSEGAPC VFPFTFLGNKYESCTSAGRSDGKMWCATTANYDDDRKWGFCPDQGYSLFLVAAHEFGHA MGLEHSQDPGALMAPIYTYTKNFRLSQDDIKGIQELYGASPDIDLGTGPTPTLGPVTPEICK QDIVFDGIAQIRGEIFFFKDRFIWRTVTPRDKPMGPLLVATFWPELPEKIDAVYEAPQEEKA VFFAGNEYWIYSASTLERGYPKPLTSLGLPPDVQRVDAAFNWSKNKKTYIFAGDKFWRYN EVKKKMDPGFPKLIADAWNAIPDNLDAVVDLQGGGHSYFFKGAYYLKLENQSLKSVKFG SIKSDWLGC (SEQ ID NO: 3313), or a fragment thereof, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) to the amino acid sequence of SEQ ID NO: 3313.

Linkers

The multispecific or multifunctional molecule disclosed herein can further include a linker, e.g., a linker between one or more of: the antigen binding domain and the cytokine molecule, the antigen binding domain and the immune cell engager, the antigen binding domain and the stromal modifying moiety, the cytokine molecule and the immune cell engager, the cytokine molecule and the stromal modifying moiety, the immune cell engager and the stromal modifying moiety, the antigen binding domain and the immunoglobulin chain constant region, the cytokine molecule and the immunoglobulin chain constant region, the immune cell engager and the immunoglobulin chain constant region, or the stromal modifying moiety and the immunoglobulin chain constant region. In embodiments, the linker is chosen from: a cleavable linker, a non-cleavable linker, a peptide linker, a flexible linker, a rigid linker, a helical linker, or a non-helical linker, or a combination thereof.

In one embodiment, the multispecific molecule can include one, two, three or four linkers, e.g., a peptide linker. In one embodiment, the peptide linker includes Gly and Ser. In some embodiments, the peptide linker is selected from GGGGS (SEQ ID NO: 3307); GGGGSGGGGS (SEQ ID NO: 3308); GGGGSGGGGSGGGGS (SEQ ID NO: 3309); and DVPSGPGGGGGSGGGGS (SEQ ID NO: 3310). In some embodiments, the peptide linker is a A(EAAAK)nA (SEQ ID NO: 3437) family of linkers (e.g., as described in Protein Eng. (2001) 14 (8): 529-532). These are stiff helical linkers with n ranging from 2-5. In some embodiments, the peptide linker is selected from AEAAAKEAAAKAAA (SEQ ID NO: 3314); AEAAAKEAAAKEAAAKAAA (SEQ ID NO: 3315); AEAAAKEAAAKEAAAKEAAAKAAA (SEQ ID NO: 3316); and AEAAAKEAAAKEAAAKEAAAKEAAAKAAA(SEQ ID NO: 3317).

Nucleic Acids

Nucleic acids encoding the aforementioned antibody molecules, e.g., anti-TCRβV antibody molecules, multispecific or multifunctional molecules are also disclosed.

In certain embodiments, the invention features nucleic acids comprising nucleotide sequences that encode heavy and light chain variable regions and CDRs or hypervariable loops of the antibody molecules, as described herein. For example, the invention features a first and second nucleic acid encoding heavy and light chain variable regions, respectively, of an antibody molecule chosen from one or more of the antibody molecules disclosed herein. The nucleic acid can comprise a nucleotide sequence as set forth in the tables herein, or a sequence substantially identical thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, or which differs by no more than 3, 6, 15, 30, or 45 nucleotides from the sequences shown in the tables herein.

In certain embodiments, the nucleic acid can comprise a nucleotide sequence encoding at least one, two, or three CDRs or hypervariable loops from a heavy chain variable region having an amino acid sequence as set forth in the tables herein, or a sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or having one or more substitutions, e.g., conserved substitutions). In other embodiments, the nucleic acid can comprise a nucleotide sequence encoding at least one, two, or three CDRs or hypervariable loops from a light chain variable region having an amino acid sequence as set forth in the tables herein, or a sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or having one or more substitutions, e.g., conserved substitutions). In yet another embodiment, the nucleic acid can comprise a nucleotide sequence encoding at least one, two, three, four, five, or six CDRs or hypervariable loops from heavy and light chain variable regions having an amino acid sequence as set forth in the tables herein, or a sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or having one or more substitutions, e.g., conserved substitutions).

In certain embodiments, the nucleic acid can comprise a nucleotide sequence encoding at least one, two, or three CDRs or hypervariable loops from a heavy chain variable region having the nucleotide sequence as set forth in the tables herein, a sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein). In another embodiment, the nucleic acid can comprise a nucleotide sequence encoding at least one, two, or three CDRs or hypervariable loops from a light chain variable region having the nucleotide sequence as set forth in the tables herein, or a sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein). In yet another embodiment, the nucleic acid can comprise a nucleotide sequence encoding at least one, two, three, four, five, or six CDRs or hypervariable loops from heavy and light chain variable regions having the nucleotide sequence as set forth in the tables herein, or a sequence substantially homologous thereto (e.g., a sequence at least about 85%, 90%, 95%, 99% or more identical thereto, and/or capable of hybridizing under the stringency conditions described herein).

In certain embodiments, the nucleic acid can comprise a nucleotide sequence encoding a cytokine molecule, an immune cell engager, or a stromal modifying moiety disclosed herein.

In another aspect, the application features host cells and vectors containing the nucleic acids described herein. The nucleic acids may be present in a single vector or separate vectors present in the same host cell or separate host cell, as described in more detail hereinbelow.

Vectors

Further provided herein are vectors comprising the nucleotide sequences encoding antibody molecules, e.g., anti-TCRβV antibody molecules, or a multispecific or multifunctional molecule described herein. In one embodiment, the vectors comprise nucleic acid sequences encoding antibody molecules, e.g., anti-TCRβV antibody molecules, or multispecific or multifunctional molecule described herein. In one embodiment, the vectors comprise the nucleotide sequences described herein. The vectors include, but are not limited to, a virus, plasmid, cosmid, lambda phage or a yeast artificial chromosome (YAC).

Numerous vector systems can be employed. For example, one class of vectors utilizes DNA elements which are derived from animal viruses such as, for example, bovine papilloma virus, polyoma virus, adenovirus, vaccinia virus, baculovirus, retroviruses (Rous Sarcoma Virus, MMTV or MOMLV) or SV40 virus. Another class of vectors utilizes RNA elements derived from RNA viruses such as Semliki Forest virus, Eastern Equine Encephalitis virus and Flaviviruses.

Additionally, cells which have stably integrated the DNA into their chromosomes may be selected by introducing one or more markers which allow for the selection of transfected host cells. The marker may provide, for example, prototropy to an auxotrophic host, biocide resistance (e.g., antibiotics), or resistance to heavy metals such as copper, or the like. The selectable marker gene can be either directly linked to the DNA sequences to be expressed, or introduced into the same cell by cotransformation. Additional elements may also be needed for optimal synthesis of mRNA. These elements may include splice signals, as well as transcriptional promoters, enhancers, and termination signals.

Once the expression vector or DNA sequence containing the constructs has been prepared for expression, the expression vectors may be transfected or introduced into an appropriate host cell. Various techniques may be employed to achieve this, such as, for example, protoplast fusion, calcium phosphate precipitation, electroporation, retroviral transduction, viral transfection, gene gun, lipid based transfection or other conventional techniques. In the case of protoplast fusion, the cells are grown in media and screened for the appropriate activity

Methods and conditions for culturing the resulting transfected cells and for recovering the antibody molecule produced are known to those skilled in the art, and may be varied or optimized depending upon the specific expression vector and mammalian host cell employed, based upon the present description.

Cells

In another aspect, the application features host cells and vectors containing the nucleic acids described herein. The nucleic acids may be present in a single vector or separate vectors present in the same host cell or separate host cell. The host cell can be a eukaryotic cell, e.g., a mammalian cell, an insect cell, a yeast cell, or a prokaryotic cell, e.g., E. coli. For example, the mammalian cell can be a cultured cell or a cell line. Exemplary mammalian cells include lymphocytic cell lines (e.g., NSO), Chinese hamster ovary cells (CHO), COS cells, oocyte cells, and cells from a transgenic animal, e.g., mammary epithelial cell.

The invention also provides host cells comprising a nucleic acid encoding an antibody molecule as described herein.

In one embodiment, the host cells are genetically engineered to comprise nucleic acids encoding the antibody molecule.

In one embodiment, the host cells are genetically engineered by using an expression cassette. The phrase “expression cassette,” refers to nucleotide sequences, which are capable of affecting expression of a gene in hosts compatible with such sequences. Such cassettes may include a promoter, an open reading frame with or without introns, and a termination signal. Additional factors necessary or helpful in effecting expression may also be used, such as, for example, an inducible promoter.

The invention also provides host cells comprising the vectors described herein.

The cell can be, but is not limited to, a eukaryotic cell, a bacterial cell, an insect cell, or a human cell. Suitable eukaryotic cells include, but are not limited to, Vero cells, HeLa cells, COS cells, CHO cells, HEK293 cells, BHK cells and MDCKII cells. Suitable insect cells include, but are not limited to, Sf9 cells.

Method of Expanding Cells with Anti-TCRVB Antibodies

Any of the compositions and methods described herein can be used to expand an immune cell population. An immune cell provided herein includes an immune cell derived from a hematopoietic stem cell or an immune cell derived from a non-hematopoietic stem cell, e.g., by differentiation or de-differentiation.

An immune cell includes a hematopoietic stem cell, progeny thereof and/or cells that have differentiated from said HSC, e.g., lymphoid cells or myeloid cells. An immune cell can be an adaptive immune cell or an innate immune cell. Examples of immune cells include T cells, B cells, Natural Killer cells, Natural Killer T cells, neutrophils, dendritic cells, monocytes, macrophages, and granulocytes.

In some embodiments of any of the methods of compositions disclosed herein, an immune cell is a T cell. In some embodiments, a T cell includes a CD4+ T cell, a CD8+ T cell, a TCR alpha-beta T cell, a TCR gamma-delta T cell. In some embodiments, a T cell comprises a memory T cell (e.g., a central memory T cell, or an effector memory T cell (e.g., a TEMRA) or an effector T cell. In some embodiments, a T cell comprises a tumor infiltrating lymphocyte (TIL).

In some embodiments of any of the methods of compositions disclosed herein, an immune cell is an NK cell.

In some embodiments of any of the methods of compositions disclosed herein, an immune cell is a TIL. TILs are immune cells (e.g., T cells, B cells or NK cells) that can be found in a tumor or around a tumor (e.g., in the stroma or tumor microenvironment of a tumor), e.g., a solid tumor, e.g., as described herein. TILs can be obtained from a sample from a subject having cancer, e.g., a biopsy or a surgical sample. In some embodiments, TILs can be expanded using a method disclosed herein. In some embodiments, a population of expanded TILs, e.g., expanded using a method disclosed herein, can be administered to a subject to treat a disease, e.g., a cancer.

In certain aspects of the present disclosure, immune cells, e.g., T cells (e.g., TILs), can be obtained from a unit of blood collected from a subject using any number of techniques known to the skilled artisan, such as Ficoll™ separation. In one aspect, cells from the circulating blood of an individual are obtained by apheresis. The apheresis product typically contains lymphocytes, including T cells, monocytes, granulocytes, B cells, other nucleated white blood cells, red blood cells, and platelets. In one aspect, the cells collected by apheresis may be washed to remove the plasma fraction and, optionally, to place the cells in an appropriate buffer or media for subsequent processing steps. In one embodiment, the cells are washed with phosphate buffered saline (PBS). In an alternative embodiment, the wash solution lacks calcium and may lack magnesium or may lack many if not all divalent cations. The methods described herein can include more than one selection step, e.g., more than one depletion step.

In one embodiment, the methods of the application can utilize culture media conditions comprising DMEM, DMEM F12, RPMI 1640, and/or AIM V media. The media can be supplemented with glutamine, HEPES buffer (e.g., 10 mM), serum (e.g., heat-inactivated serum, e.g., 10%), and/or beta mercaptoethanol (e.g., 55 uM). IN some embodiments, the culture conditions disclosed herein comprise one or more supplements, cytokines, growth factors, or hormones. In some embodiments, the culture condition comprises one or more of IL-2, IL-15, or IL-7, or a combination thereof.

Immune effector cells such as T cells may be activated and expanded generally using methods as described, for example, in U.S. Pat. Nos. 6,352,694; 6,534,055; or 6,905,680. Generally, a population of immune cells, may be expanded by contact with an agent that stimulates a CD3/TCR complex associated signal and a ligand that stimulates a costimulatory molecule on the surface of the T cells; and/or by contact with a cytokine, e.g., IL-2, IL-15 or IL-7. T cell expansion protocols can also include stimulation, such as by contact with an anti-CD3 antibody, or antigen-binding fragment thereof, or an anti-CD2 antibody immobilized on a surface, or by contact with a protein kinase C activator (e.g., bryostatin) in conjunction with a calcium ionophore. For example, a population of T cells can be contacted with an anti-CD3 antibody and an anti-CD28 antibody, under conditions appropriate for stimulating proliferation of the T cells. To stimulate proliferation of either CD4+ T cells or CD8+ T cells, an anti-CD3 antibody and an anti-CD28 antibody can be used. Examples of an anti-CD28 antibody include 9.3, B-T3, XR-CD28 (Diaclone, Besancon, France) can be used as can other methods commonly known in the art (Berg et al., Transplant Proc. 30(8):3975-3977, 1998; Haanen et al., J. Exp. Med. 190(9):13191328, 1999; Garland et al., J. Immunol Meth. 227(1-2):53-63, 1999).

A TIL population can also be expanded by methods known in the art. For example, a population of TILs can be expanded as described in Hall et al., Journal for ImmunoTherapy of Cancer (2016) 4:61, the entire contents of which are hereby incorporated by reference. Briefly, TILs can be isolated from a sample by mechanical and/or physical digestion. The resultant TIL population can be stimulated with an anti-CD3 antibody in the presence of non-dividing feeder cells. In some embodiments, the TIL population can be cultured, e.g., expanded, in the presence of IL-2, e.g., human IL-2. In some embodiments, the TIL cells can be cultured, e.g., expanded for a period of at least 1-21 days, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 days.

As disclosed herein, in some embodiments, an immune cell population (e.g., a T cell (e.g., a TEMRA cell or a TIL population) can be expanded by contacting the immune cell population with an anti-TCRVB antibody, e.g., as described herein.

In some embodiments, the expansion occurs in vivo, e.g., in a subject. In some embodiments, a subject is administered an anti-TCRβV antibody molecule disclosed herein resulting in expansion of immune cells in vivo.

In some embodiments, the expansion occurs ex vivo, e.g., in vitro. In some embodiments, cells from a subject, e.g., T cells, e.g., TIL cells, are expanded in vitro with an anti-TCRβV antibody molecule disclosed herein. In some embodiments, the expanded TILs are administered to the subject to treat a disease or a symptom of a disease.

In some embodiments, a method of expansion disclosed herein results in an expansion of at least 1.1-10 fold, 10-20 fold, or 20-50 fold expansion. In some embodiments, the expansion is at least 1.1, 1.2, 1.3, 1.4, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45 or 50 fold expansion.

In some embodiments, a method of expansion disclosed herein comprises culturing, e.g., expanding, the cells for at least about 4 hours, 6 hours, 10 hours, 12 hours, 15 hours, 18 hours, 20 hours, or 22 hours. In some embodiments, a method of expansion disclosed herein comprises culturing, e.g., expanding, the cells for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 1,6 17, 18, 19, 20 or 21 days. In some embodiments, a method of expansion disclosed herein comprises culturing, e.g., expanding, the cells for at least about 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks or 8 weeks.

In some embodiments, a method of expansion disclosed herein is performed on immune cells obtained from a healthy subject.

In some embodiments, a method of expansion disclosed herein is performed on immune cells (e.g., TILs) obtained from a subject having a disease, e.g., a cancer, e.g., a solid tumor as disclosed herein.

In some embodiments, a method of expansion disclosed herein further comprises contacting the population of cells with an agent, that promotes, e.g., increases, immune cell expansion. In some embodiments, the agent comprises an immune checkpoint inhibitor, e.g., a PD-1 inhibitor, a LAG-3 inhibitor, a CTLA4 inhibitor, or a TIM-3 inhibitor. In some embodiments, the agent comprises a 4-1BB agonist, e.g., an anti-4-1BB antibody.

Without wishing to be bound by theory, it is believed that an anti-TCRβV antibody molecule disclosed herein can expand, e.g., selectively or preferentially expand, T cells expressing a T cell receptor (TCR) comprising a TCR alpha and/or TCR beta molecule, e.g., TCR alpha-beta T cells (αβ T cells). In some embodiments, an anti-TCRβV antibody molecule disclosed herein does not expand, or induce proliferation of T cells expressing a TCR comprising a TCR gamma and/or TCR delta molecule, e.g., TCR gamma-delta T cells (γδ T cells). In some embodiments, an anti-TCRβV antibody molecule disclosed herein, selectively or preferentially expands αβ T cells over γδ T cells.

Without wishing to be bound by theory, it is believed that, in some embodiments, γδ T cells are associated with cytokine release syndrome (CRS) and/or neurotoxicity (NT). In some embodiments, an anti-TCRβV antibody molecule disclosed herein results in selective expansion of non-γδ T cells, e.g., expansion of αβ T cells, thus reducing CRS and/or NT.

In some embodiments, any of the compositions or methods disclosed herein result in an immune cell population having a reduction of, e.g., depletion of, γδ T cells. In some embodiments, the immune cell population is contacted with an agent that reduces, e.g., inhibits or depletes, γδ T cells, e.g., an anti-IL-17 antibody or an agent that binds to a TCR gamma and/or TCR delta molecule.

Uses and Combination Therapies

Methods described herein include treating a cancer in a subject by using an anti-TCRβV antibody molecule, a multispecific or multifunctional molecule described herein, e.g., using a pharmaceutical composition described herein. Also provided are methods for reducing or ameliorating a symptom of a cancer in a subject, as well as methods for inhibiting the growth of a cancer and/or killing one or more cancer cells. In embodiments, the methods described herein decrease the size of a tumor and/or decrease the number of cancer cells in a subject administered with a described herein or a pharmaceutical composition described herein.

In embodiments, the cancer is a hematological cancer. In embodiments, the hematological cancer is a leukemia or a lymphoma. As used herein, a “hematologic cancer” refers to a tumor of the hematopoietic or lymphoid tissues, e.g., a tumor that affects blood, bone marrow, or lymph nodes. Exemplary hematologic malignancies include, but are not limited to, leukemia (e.g., acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), hairy cell leukemia, acute monocytic leukemia (AMoL), chronic myelomonocytic leukemia (CMML), juvenile myelomonocytic leukemia (JMML), or large granular lymphocytic leukemia), lymphoma (e.g., AIDS-related lymphoma, cutaneous T-cell lymphoma, Hodgkin lymphoma (e.g., classical Hodgkin lymphoma or nodular lymphocyte-predominant Hodgkin lymphoma), mycosis fungoides, non-Hodgkin lymphoma (e.g., B-cell non-Hodgkin lymphoma (e.g., Burkitt lymphoma, small lymphocytic lymphoma (CLL/SLL), diffuse large B-cell lymphoma, follicular lymphoma, immunoblastic large cell lymphoma, precursor B-lymphoblastic lymphoma, or mantle cell lymphoma) or T-cell non-Hodgkin lymphoma (mycosis fungoides, anaplastic large cell lymphoma, or precursor T-lymphoblastic lymphoma)), primary central nervous system lymphoma, Sézary syndrome, Waldenström macroglobulinemia), chronic myeloproliferative neoplasm, Langerhans cell histiocytosis, multiple myeloma/plasma cell neoplasm, myelodysplastic syndrome, or myelodysplastic/myeloproliferative neoplasm.

In embodiments, the cancer is a myeloproliferative neoplasm, e.g., primary or idiopathic myelofibrosis (MF), essential thrombocytosis (ET), polycythemia vera (PV), or chronic myelogenous leukemia (CIVIL). In embodiments, the cancer is myelofibrosis. In embodiments, the subject has myelofibrosis. In embodiments, the subject has a calreticulin mutation, e.g., a calreticulin mutation disclosed herein. In embodiments, the subject does not have the JAK2-V617F mutation. In embodiments, the subject has the JAK2-V617F mutation. In embodiments, the subject has a MPL mutation. In embodiments, the subject does not have a MPL mutation.

In embodiments, the cancer is a solid cancer. Exemplary solid cancers include, but are not limited to, ovarian cancer, rectal cancer, stomach cancer, testicular cancer, cancer of the anal region, uterine cancer, colon cancer, rectal cancer, renal-cell carcinoma, liver cancer, non-small cell carcinoma of the lung, cancer of the small intestine, cancer of the esophagus, melanoma, Kaposi's sarcoma, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular malignant melanoma, uterine cancer, brain stem glioma, pituitary adenoma, epidermoid cancer, carcinoma of the cervix squamous cell cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the vagina, sarcoma of soft tissue, cancer of the urethra, carcinoma of the vulva, cancer of the penis, cancer of the bladder, cancer of the kidney or ureter, carcinoma of the renal pelvis, spinal axis tumor, neoplasm of the central nervous system (CNS), primary CNS lymphoma, tumor angiogenesis, metastatic lesions of said cancers, or combinations thereof.

In some embodiments, the cancer is acute lymphoblastic leukemia, acute lymphocytic leukemia, acute myelogenous leukemia, aplastic anemia, chronic myelogenous leukemia, desmoplastic small round cell tumor, Ewing's sarcoma, Hodgkin's disease, multiple myeloma, myelodysplasia, Non-Hodgkin's lymphoma, paroxysmal nocturnal hemoglobinuria, radiation poisoning, chronic lymphocytic leukemia, AL amyloidosis, essential thrombocytosis, polycythemia vera, severe aplastic anemia, neuroblastoma, breast tumors, ovarian tumors, renal cell carcinoma, autoimmune disorders, such as systemic sclerosis, osteopetrosis, inherited metabolic disorders, juvenile chronic arthritis, adrenoleukodystrophy, amegakaryocytic thrombocytopenia, sickle cell disease, severe congenital immunodeficiency, Griscelli syndrome type II, Hurler syndrome, Kostmann syndrome, Krabbe disease, metachromatic leukodystrophy, thalassemia, hemophagocytic lymphohistiocytosis, and Wiskott-Aldrich syndrome, leukemias, lymphomas, melanomas, neuroendocrine tumors, carcinomas and sarcomas. Exemplary cancers that may be treated with a compound, pharmaceutical composition, or method provided herein include lymphoma, sarcoma, bladder cancer, bone cancer, brain tumor, cervical cancer, colon cancer, esophageal cancer, gastric cancer, head and neck cancer, kidney cancer, myeloma, thyroid cancer, leukemia, prostate cancer, breast cancer (e.g. triple negative, ER positive, ER negative, chemotherapy resistant, herceptin resistant, HER2 positive, doxorubicin resistant, tamoxifen resistant, ductal carcinoma, lobular carcinoma, primary, metastatic), ovarian cancer, pancreatic cancer, liver cancer (e.g., hepatocellular carcinoma), lung cancer (e.g. non-small cell lung carcinoma, squamous cell lung carcinoma, adenocarcinoma, large cell lung carcinoma, small cell lung carcinoma, carcinoid, sarcoma), glioblastoma multiforme, glioma, melanoma, prostate cancer, castration-resistant prostate cancer, breast cancer, triple negative breast cancer, glioblastoma, ovarian cancer, lung cancer, squamous cell carcinoma (e.g., head, neck, or esophagus), colorectal cancer, leukemia, acute myeloid leukemia, lymphoma, B cell lymphoma, or multiple myeloma. Additional examples include, cancer of the thyroid, endocrine system, brain, breast, cervix, colon, head & neck, esophagus, liver, kidney, lung, non-small cell lung, melanoma, mesothelioma, ovary, sarcoma, stomach, uterus or Medulloblastoma, Hodgkin's Disease, Non-Hodgkin's Lymphoma, multiple myeloma, neuroblastoma, glioma, glioblastoma multiforme, ovarian cancer, rhabdomyosarcoma, primary thrombocytosis, primary macroglobulinemia, primary brain tumors, cancer, malignant pancreatic insulanoma, malignant carcinoid, urinary bladder cancer, premalignant skin lesions, testicular cancer, lymphomas, thyroid cancer, neuroblastoma, esophageal cancer, genitourinary tract cancer, malignant hypercalcemia, endometrial cancer, adrenal cortical cancer, neoplasms of the endocrine or exocrine pancreas, medullary thyroid cancer, medullary thyroid carcinoma, melanoma, colorectal cancer, papillary thyroid cancer, hepatocellular carcinoma, Paget's Disease of the Nipple, Phyllodes Tumors, Lobular Carcinoma, Ductal Carcinoma, cancer of the pancreatic stellate cells, cancer of the hepatic stellate cells, or prostate cancer. In some embodiments, the cancer is a solid tumor. In some embodiments, the cancer is hematological.

In embodiments, the anti-TCRβV antibody molecule, multispecific or multifunctional molecules (or pharmaceutical composition) are administered in a manner appropriate to the disease to be treated or prevented. The quantity and frequency of administration will be determined by such factors as the condition of the patient, and the type and severity of the patient's disease. Appropriate dosages may be determined by clinical trials. For example, when “an effective amount” or “a therapeutic amount” is indicated, the precise amount of the pharmaceutical composition (or multispecific or multifunctional molecules) to be administered can be determined by a physician with consideration of individual differences in tumor size, extent of infection or metastasis, age, weight, and condition of the subject. In embodiments, the pharmaceutical composition described herein can be administered at a dosage of 104 to 109 cells/kg body weight, e.g., 105 to 106 cells/kg body weight, including all integer values within those ranges. In embodiments, the pharmaceutical composition described herein can be administered multiple times at these dosages. In embodiments, the pharmaceutical composition described herein can be administered using infusion techniques described in immunotherapy (see, e.g., Rosenberg et al., New Eng. J. of Med. 319:1676, 1988).

In embodiments, the anti-TCRβV antibody molecule, multispecific or multifunctional molecules or pharmaceutical composition is administered to the subject parentally. In embodiments, the cells are administered to the subject intravenously, subcutaneously, intratumorally, intranodally, intramuscularly, intradermally, or intraperitoneally. In embodiments, the cells are administered, e.g., injected, directly into a tumor or lymph node. In embodiments, the cells are administered as an infusion (e.g., as described in Rosenberg et al., New Eng. J. of Med. 319:1676, 1988) or an intravenous push. In embodiments, the cells are administered as an injectable depot formulation.

In embodiments, the subject is a mammal. In embodiments, the subject is a human, monkey, pig, dog, cat, cow, sheep, goat, rabbit, rat, or mouse. In embodiments, the subject is a human. In embodiments, the subject is a pediatric subject, e.g., less than 18 years of age, e.g., less than 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1 or less years of age. In embodiments, the subject is an adult, e.g., at least 18 years of age, e.g., at least 19, 20, 21, 22, 23, 24, 25, 25-30, 30-35, 35-40, 40-50, 50-60, 60-70, 70-80, or 80-90 years of age.

Methods of Cancer Treatment

Methods described herein include treating a cancer in a subject by using an anti-TCRβV antibody molecule, e.g., using a pharmaceutical composition described herein. Also provided are methods for reducing or ameliorating a symptom of a cancer in a subject, as well as methods for inhibiting the growth of a cancer and/or killing one or more cancer cells. In embodiments, the methods described herein decrease the size of a tumor and/or decrease the number of cancer cells in a subject administered with a described herein or a pharmaceutical composition described herein.

Disclosed herein are methods of treating a subject having a cancer comprising acquiring a status of one or more TCRBV molecules in a subject. In some embodiments, a higher, e.g., increased, level or activity of one or more TCRβV molecules in a subject, e.g., in a sample from a subject, is indicative of a bias, e.g., a preferential expansion, e.g., clonal expansion, of T cells expressing said one or more TCRβV molecules in the subject.

Without wishing to be bound by theory, it is believed that a biased T cell population, e.g., a T cell population expressing a TCRβV molecule, is antigen-specific for a disease antigen, e.g., a cancer antigen (Wang C Y, et al., Int J Oncol. (2016) 48(6):2247-56). In some embodiments, the cancer antigen comprises a cancer associated antigen or a neoantigen. In some embodiments, a subject having a cancer, e.g., as disclosed herein, has a higher, e.g., increased, level or activity of one or more TCRβV molecules associated with the cancer. In some embodiments, the TCRβV molecule is associated with, e.g., recognizes, a cancer antigen, e.g., a cancer associated antigen or a neoantigen.

Accordingly, disclosed herein are methods of expanding an immune effector cell population obtained from a subject, comprising acquiring a status of one or more TCRβV molecules in a sample from the subject, comprising contacting said immune effector cell population with an anti-TCRβV antibody molecule disclosed herein, e.g., an anti-TCRβV antibody molecule that binds to the same TCRβV molecule that is higher, e.g., increased in the immune effector cell population in the sample from the subject. In some embodiments, contacting the population of immune effector cells (e.g., comprising T cells that express one or more TCRβV molecules) with an anti-TCRβV molecule results in expansion of the population of immune effector cells expressing one or more TCRβV molecules. In some embodiments, the expanded population, or a portion thereof, is administered to the subject (e.g., same subject from whom the immune effector cell population was obtained), to treat the cancer. In some embodiments, the expanded population, or a portion thereof, is administered to a different subject (e.g., not the same subject from whom the immune effector cell population was obtained), to treat the cancer.

Also disclosed herein, are methods of treating a subject having a cancer, comprising: acquiring a status of one or more TCRβV molecules in a sample from the subject, and determining whether the one or more TCRβV molecules is higher, e.g., increased, in a sample from the subject compared to a reference value, wherein responsive to said determination, administering to the subject an effective amount of an anti-TCRβV antibody molecule, e.g., an agonistic anti-TCRβV antibody molecule, e.g., as described herein.

In some embodiments of any of the methods or composition for use disclosed herein, the subject has B-CLL. In some embodiments, a subject having B-CLL has a higher, e.g., increased, level or activity of one or more TCRβV molecules, e.g., one or more TCRβV molecules comprising: (i) TCRβ V6 subfamily comprising, e.g., TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01; (ii) TCRβ V5 subfamily comprising TCRβ V5-6*01, TCRβ V5-4*01, or TCRβ V5-8*01; (iii) TCRβ V3 subfamily comprising TCRβ V3-1*01; (iv) TCRβ V2 subfamily comprising TCRβ V2*01; or (v) TCRβ V19 subfamily comprising TCRβ V19*01, or TCRβ V19*02.

In some embodiments, a subject having B-CLL has a higher, e.g., increased, level or activity of a TCRβ V6 subfamily comprising, e.g., TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V6 subfamily. In some embodiments, administration of the an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V6 subfamily.

In some embodiments, a subject having B-CLL has a higher, e.g., increased, level or activity of a TCRβ V5 subfamily comprising TCRβ V5-6*01, TCRβ V5-4*01, or TCRβ V5-8*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V5 subfamily. In some embodiments, administration of the an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V5 subfamily.

In some embodiments, a subject having B-CLL has a higher, e.g., increased, level or activity of a TCRβ V3 subfamily comprising TCRβ V3-1*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V3 subfamily. In some embodiments, administration of the an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V3 subfamily.

In some embodiments, a subject having B-CLL has a higher, e.g., increased, level or activity of a TCRβ V2 subfamily comprising TCRβ V2*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V2 subfamily. In some embodiments, administration of the an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V2 subfamily.

In some embodiments, a subject having B-CLL has a higher, e.g., increased, level or activity of a TCRβ V19 subfamily comprising TCRβ V19*01, or TCRβ V19*02. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRBV molecule as described herein) that binds to one or more members of the TCRβ V19 subfamily. In some embodiments, administration of the an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V19 subfamily.

In some embodiments of any of the methods or composition for use disclosed herein, the subject has melanoma. In some embodiments, a subject having melanoma has a higher, e.g., increased, level or activity of one or more TCRβV molecules, e.g., one or more TCRβV molecules comprising the TCRβ V6 subfamily comprising, e.g., TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V6 subfamily. In some embodiments, administration of the an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V6 subfamily.

In some embodiments of any of the methods or composition for use disclosed herein, the subject has DLBCL. In some embodiments, a subject having melanoma has a higher, e.g., increased, level or activity of one or more TCRβV molecules, e.g., one or more TCRβV molecules comprising: (i) TCRβ V13 subfamily comprising TCRβ V13*01; (ii) TCRβ V3 subfamily comprising TCRβ V3-1*01; or (iii) TCRβ V23 subfamily.

In some embodiments, a subject having DLBCL has a higher, e.g., increased, level or activity of a TCRβ V13 subfamily comprising TCRβ V13*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V13 subfamily. In some embodiments, administration of the an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V13 subfamily.

In some embodiments, a subject having DLBCL has a higher, e.g., increased, level or activity of a TCRβ V3 subfamily comprising TCRβ V3-1*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V3 subfamily. In some embodiments, administration of the an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V3 subfamily.

In some embodiments, a subject having DLBCL has a higher, e.g., increased, level or activity of a TCRβ V23 subfamily. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V23 subfamily. In some embodiments, administration of the an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V23 subfamily.

In some embodiments of any of the methods or composition for use disclosed herein, the subject has CRC. In some embodiments, a subject having melanoma has a higher, e.g., increased, level or activity of one or more TCRβV molecules, e.g., one or more TCRβV molecules comprising: (i) TCRβ V19 subfamily comprising TCRβ V19*01, or TCRβ V19*02; (ii) TCRβ V12 subfamily comprising TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01; (iii) TCRβ V16 subfamily comprising TCRβ V16*01; or (iv) TCRβ V21 subfamily.

In some embodiments, a subject having CRC has a higher, e.g., increased, level or activity of a TCRβ V19 subfamily comprising TCRβ V19*01, or TCRβ V19*02. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V19 subfamily. In some embodiments, administration of the an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V19 subfamily.

In some embodiments, a subject having CRC has a higher, e.g., increased, level or activity of a TCRβ V12 subfamily comprising TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V12 subfamily. In some embodiments, administration of the an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V12 subfamily.

In some embodiments, a subject having CRC has a higher, e.g., increased, level or activity of a TCRβ V16 subfamily comprising TCRβ V16*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V16 subfamily. In some embodiments, administration of the an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V16 subfamily.

In some embodiments, a subject having CRC has a higher, e.g., increased, level or activity of a TCRβ V21 subfamily. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V21 subfamily. In some embodiments, administration of the an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V21 subfamily.

In some embodiments, acquiring a value for the status, e.g., presence, level and/or activity, of one or more TCRβV molecules comprises acquiring a measure of the T cell receptor (TCR) repertoire of a sample. In some embodiments, the value comprises a measure of the clonotype of a population of T cells in the sample.

In some embodiments, a value for the status of one or more TCRβV molecules is obtained, e.g., measured, using an assay described in Wang C Y, et al., Int J Oncol. (2016) 48(6):2247-56, the entire contents of which are hereby incorporated by reference.

In some embodiments, a value for the status of one or more TCRβV molecules is obtained, e.g., measured, using flow cytometry.

Combination Therapies

The anti-TCRβV antibody molecule, multispecific or multifunctional molecules disclosed herein can be used in combination with a second therapeutic agent or procedure.

In embodiments, the anti-TCRβV antibody molecule, multispecific or multifunctional molecule and the second therapeutic agent or procedure are administered/performed after a subject has been diagnosed with a cancer, e.g., before the cancer has been eliminated from the subject. In embodiments, the anti-TCRβV antibody molecule, multispecific or multifunctional molecule and the second therapeutic agent or procedure are administered/performed simultaneously or concurrently. For example, the delivery of one treatment is still occurring when the delivery of the second commences, e.g., there is an overlap in administration of the treatments. In other embodiments, the anti-TCRβV antibody molecule, multispecific or multifunctional molecule and the second therapeutic agent or procedure are administered/performed sequentially. For example, the delivery of one treatment ceases before the delivery of the other treatment begins.

In embodiments, combination therapy can lead to more effective treatment than monotherapy with either agent alone. In embodiments, the combination of the first and second treatment is more effective (e.g., leads to a greater reduction in symptoms and/or cancer cells) than the first or second treatment alone. In embodiments, the combination therapy permits use of a lower dose of the first or the second treatment compared to the dose of the first or second treatment normally required to achieve similar effects when administered as a monotherapy. In embodiments, the combination therapy has a partially additive effect, wholly additive effect, or greater than additive effect.

In one embodiment, the anti-TCRBV antibody, multispecific or multifunctional molecule is administered in combination with a therapy, e.g., a cancer therapy (e.g., one or more of anti-cancer agents, immunotherapy, photodynamic therapy (PDT), surgery and/or radiation). The terms “chemotherapeutic,” “chemotherapeutic agent,” and “anti-cancer agent” are used interchangeably herein. The administration of the multispecific or multifunctional molecule and the therapy, e.g., the cancer therapy, can be sequential (with or without overlap) or simultaneous. Administration of the anti-TCRBV antibody, multispecific or multifunctional molecule can be continuous or intermittent during the course of therapy (e.g., cancer therapy). Certain therapies described herein can be used to treat cancers and non-cancerous diseases. For example, PDT efficacy can be enhanced in cancerous and non-cancerous conditions (e.g., tuberculosis) using the methods and compositions described herein (reviewed in, e.g., Agostinis, P. et al. (2011) CA Cancer J. Clin. 61:250-281).

Anti-Cancer Therapies

In other embodiments, the anti-TCRβV antibody molecule, multispecific or multifunctional molecule is administered in combination with a low or small molecular weight chemotherapeutic agent. Exemplary low or small molecular weight chemotherapeutic agents include, but not limited to, 13-cis-retinoic acid (isotretinoin, ACCUTANE®), 2-CdA (2-chlorodeoxyadenosine, cladribine, LEUSTATIN™), 5-azacitidine (azacitidine, VIDAZA®), 5-fluorouracil (5-FU, fluorouracil, ADRUCIL®), 6-mercaptopurine (6-MP, mercaptopurine, PURINETHOL®), 6-TG (6-thioguanine, thioguanine, THIOGUANINE TABLOID®), abraxane (paclitaxel protein-bound), actinomycin-D (dactinomycin, COSMEGEN®), alitretinoin (PANRETIN®), all-transretinoic acid (ATRA, tretinoin, VESANOID®), altretamine (hexamethylmelamine, HMM, HEXALEN®), amethopterin (methotrexate, methotrexate sodium, MTX, TREXALL™, RHEUMATREX®), amifostine (ETHYOL®), arabinosylcytosine (Ara-C, cytarabine, CYTOSAR-U®), arsenic trioxide (TRISENOX®), asparaginase (Erwinia L-asparaginase, L-asparaginase, ELSPAR®, KIDROLASE®), BCNU (carmustine, BiCNU®), bendamustine (TREANDA®), bexarotene (TARGRETIN®), bleomycin (BLENOXANE®), busulfan (BUSULFEX®, MYLERAN®), calcium leucovorin (Citrovorum Factor, folinic acid, leucovorin), camptothecin-11 (CPT-11, irinotecan, CAMPTOSAR®), capecitabine (XELODA®), carboplatin (PARAPLATIN®), carmustine wafer (prolifeprospan 20 with carmustine implant, GLIADEL® wafer), CCI-779 (temsirolimus, TORISEL®), CCNU (lomustine, CeeNU), CDDP (cisplatin, PLATINOL®, PLATINOL-AQ®), chlorambucil (leukeran), cyclophosphamide (CYTOXAN®, NEOSAR®), dacarbazine (DIC, DTIC, imidazole carboxamide, DTIC-DOME®), daunomycin (daunorubicin, daunorubicin hydrochloride, rubidomycin hydrochloride, CERUBIDINE®), decitabine (DACOGEN®), dexrazoxane (ZINECARD®), DHAD (mitoxantrone, NOVANTRONE®), docetaxel (TAXOTERE®), doxorubicin (ADRIAMYCIN®, RUBEX®), epirubicin (ELLENCE™), estramustine (EMCYT®), etoposide (VP-16, etoposide phosphate, TOPOSAR®, VEPESID®, ETOPOPHOS®), floxuridine (FUDR®), fludarabine (FLUDARA®), fluorouracil (cream) (CARAC™, EFUDEX®, FLUOROPLEX®), gemcitabine (GEMZAR®), hydroxyurea (HYDREA®, DROXIA™, MYLOCEL™), idarubicin (IDAMYCIN®), ifosfamide (IFEX®), ixabepilone (IXEMPRA™), LCR (leurocristine, vincristine, VCR, ONCOVIN®, VINCASAR PFS®), L-PAM (L-sarcolysin, melphalan, phenylalanine mustard, ALKERAN®), mechlorethamine (mechlorethamine hydrochloride, mustine, nitrogen mustard, MUSTARGEN®), mesna (MESNEX™), mitomycin (mitomycin-C, MTC, MUTAMYCIN®), nelarabine (ARRANON®), oxaliplatin (ELOXATIN™), paclitaxel (TAXOL®, ONXAL™), pegaspargase (PEG-L-asparaginase, ONCOSPAR®), PEMETREXED (ALIMTA®), pentostatin (NIPENT®), procarbazine (MATULANE®), streptozocin (ZANOSAR®), temozolomide (TEMODAR®), teniposide (VM-26, VUMON®), TESPA (thiophosphoamide, thiotepa, TSPA, THIOPLEX®), topotecan (HYCAMTIN®), vinblastine (vinblastine sulfate, vincaleukoblastine, VLB, ALKABAN-AQ®, VELBAN®), vinorelbine (vinorelbine tartrate, NAVELBINE®), and vorinostat (ZOLINZA®).

In another embodiment, the anti-TCRβV antibody molecule, multispecific or multifunctional molecule is administered in conjunction with a biologic. Biologics useful in the treatment of cancers are known in the art and a binding molecule of the invention may be administered, for example, in conjunction with such known biologics. For example, the FDA has approved the following biologics for the treatment of breast cancer: HERCEPTIN® (trastuzumab, Genentech Inc., South San Francisco, Calif.; a humanized monoclonal antibody that has anti-tumor activity in HER2-positive breast cancer); FASLODEX® (fulvestrant, AstraZeneca Pharmaceuticals, LP, Wilmington, Del.; an estrogen-receptor antagonist used to treat breast cancer); ARIMIDEX® (anastrozole, AstraZeneca Pharmaceuticals, LP; a nonsteroidal aromatase inhibitor which blocks aromatase, an enzyme needed to make estrogen); Aromasin® (exemestane, Pfizer Inc., New York, N.Y.; an irreversible, steroidal aromatase inactivator used in the treatment of breast cancer); FEMARA® (letrozole, Novartis Pharmaceuticals, East Hanover, N.J.; a nonsteroidal aromatase inhibitor approved by the FDA to treat breast cancer); and NOLVADEX® (tamoxifen, AstraZeneca Pharmaceuticals, LP; a nonsteroidal antiestrogen approved by the FDA to treat breast cancer). Other biologics with which the binding molecules of the invention may be combined include: AVASTIN® (bevacizumab, Genentech Inc.; the first FDA-approved therapy designed to inhibit angiogenesis); and ZEVALIN® (ibritumomab tiuxetan, Biogen Idec, Cambridge, Mass.; a radiolabeled monoclonal antibody currently approved for the treatment of B-cell lymphomas).

In addition, the FDA has approved the following biologics for the treatment of colorectal cancer: AVASTIN®; ERBITUX® (cetuximab, ImClone Systems Inc., New York, N.Y., and Bristol-Myers Squibb, New York, N.Y.; is a monoclonal antibody directed against the epidermal growth factor receptor (EGFR)); GLEEVEC® (imatinib mesylate; a protein kinase inhibitor); and ERGAMISOL® (levamisole hydrochloride, Janssen Pharmaceutica Products, LP, Titusville, N.J.; an immunomodulator approved by the FDA in 1990 as an adjuvant treatment in combination with 5-fluorouracil after surgical resection in patients with Dukes' Stage C colon cancer).

For the treatment of lung cancer, exemplary biologics include TARCEVA® (erlotinib HCL, OSI Pharmaceuticals Inc., Melville, N.Y.; a small molecule designed to target the human epidermal growth factor receptor 1 (HER1) pathway).

For the treatment of multiple myeloma, exemplary biologics include VELCADE® (bortezomib, Millennium Pharmaceuticals, Cambridge Mass.; a proteasome inhibitor). Additional biologics include THALIDOMID® (thalidomide, Clegene Corporation, Warren, N.J.; an immunomodulatory agent and appears to have multiple actions, including the ability to inhibit the growth and survival of myeloma cells and anti-angiogenesis).

Additional exemplary cancer therapeutic antibodies include, but are not limited to, 3F8, abagovomab, adecatumumab, afutuzumab, alacizumab pegol, alemtuzumab (CAMPATH®, MABCAMPATH®), altumomab pentetate (HYBRI-CEAKER®), anatumomab mafenatox, anrukinzumab (IMA-638), apolizumab, arcitumomab (CEA-SCAN®), bavituximab, bectumomab (LYMPHOSCAN®), belimumab (BENLYSTA®, LYMPHOSTAT-B®), besilesomab (SCINTIMUN®), bevacizumab (AVASTIN®), bivatuzumab mertansine, blinatumomab, brentuximab vedotin, cantuzumab mertansine, capromab pendetide (PROSTASCINT®), catumaxomab (REMOVAB®), CC49, cetuximab (C225, ERBITUX®), citatuzumab bogatox, cixutumumab, clivatuzumab tetraxetan, conatumumab, dacetuzumab, denosumab (PROLIA®), detumomab, ecromeximab, edrecolomab (PANOREX®), elotuzumab, epitumomab cituxetan, epratuzumab, ertumaxomab (REXOMUN®), etaracizumab, farletuzumab, figitumumab, fresolimumab, galiximab, gemtuzumab ozogamicin (MYLOTARG®), girentuximab, glembatumumab vedotin, ibritumomab (ibritumomab tiuxetan, ZEVALIN®), igovomab (INDIMACIS-125®), intetumumab, inotuzumab ozogamicin, ipilimumab, iratumumab, labetuzumab (CEA-CIDE®), lexatumumab, lintuzumab, lucatumumab, lumiliximab, mapatumumab, matuzumab, milatuzumab, minretumomab, mitumomab, nacolomab tafenatox, naptumomab estafenatox, necitumumab, nimotuzumab (THERACIM®, THERALOC®), nofetumomab merpentan (VERLUMA®), ofatumumab (ARZERRA®), olaratumab, oportuzumab monatox, oregovomab (OVAREX®), panitumumab (VECTIBIX®), pemtumomab (THERAGYN®), pertuzumab (OMNITARG®), pintumomab, pritumumab, ramucirumab, ranibizumab (LUCENTIS®), rilotumumab, rituximab (MABTHERA®, RITUXAN®), robatumumab, satumomab pendetide, sibrotuzumab, siltuximab, sontuzumab, tacatuzumab tetraxetan (AFP-CIDE®), taplitumomab paptox, tenatumomab, TGN1412, ticilimumab (tremelimumab), tigatuzumab, TNX-650, tositumomab (BEXXAR®), trastuzumab (HERCEPTIN®), tremelimumab, tucotuzumab celmoleukin, veltuzumab, volociximab, votumumab (HUMASPECT®), zalutumumab (HUMAX-EGFR®), and zanolimumab (HUMAX-CD4®).

In other embodiments, the anti-TCRβV antibody molecule, multispecific or multifunctional molecule is administered in combination with a viral cancer therapeutic agent. Exemplary viral cancer therapeutic agents include, but not limited to, vaccinia virus (vvDD-CDSR), carcinoembryonic antigen-expressing measles virus, recombinant vaccinia virus (TK-deletion plus GM-CSF), Seneca Valley virus-001, Newcastle virus, coxsackie virus A21, GL-ONC1, EBNA1 C-terminal/LMP2 chimeric protein-expressing recombinant modified vaccinia Ankara vaccine, carcinoembryonic antigen-expressing measles virus, G207 oncolytic virus, modified vaccinia virus Ankara vaccine expressing p53, OncoVEX GM-CSF modified herpes-simplex 1 virus, fowlpox virus vaccine vector, recombinant vaccinia prostate-specific antigen vaccine, human papillomavirus 16/18 L1 virus-like particle/AS04 vaccine, MVA-EBNA1/LMP2 Inj. vaccine, quadrivalent HPV vaccine, quadrivalent human papillomavirus (types 6, 11, 16, 18) recombinant vaccine (GARDASIL®), recombinant fowlpox-CEA(6D)/TRICOM vaccine; recombinant vaccinia-CEA(6D)-TRICOM vaccine, recombinant modified vaccinia Ankara-5T4 vaccine, recombinant fowlpox-TRICOM vaccine, oncolytic herpes virus NV1020, HPV L1 VLP vaccine V504, human papillomavirus bivalent (types 16 and 18) vaccine (CERVARIX®), herpes simplex virus HF10, Ad5CMV-p53 gene, recombinant vaccinia DF3/MUC1 vaccine, recombinant vaccinia-MUC-1 vaccine, recombinant vaccinia-TRICOM vaccine, ALVAC MART-1 vaccine, replication-defective herpes simplex virus type I (HSV-1) vector expressing human Preproenkephalin (NP2), wild-type reovirus, reovirus type 3 Dearing (REOLYSIN®), oncolytic virus HSV1716, recombinant modified vaccinia Ankara (MVA)-based vaccine encoding Epstein-Barr virus target antigens, recombinant fowlpox-prostate specific antigen vaccine, recombinant vaccinia prostate-specific antigen vaccine, recombinant vaccinia-B7.1 vaccine, rAd-p53 gene, Ad5-delta24RGD, HPV vaccine 580299, JX-594 (thymidine kinase-deleted vaccinia virus plus GM-CSF), HPV-16/18 L1/AS04, fowlpox virus vaccine vector, vaccinia-tyrosinase vaccine, MEDI-517 HPV-16/18 VLP ASO4 vaccine, adenoviral vector containing the thymidine kinase of herpes simplex virus TK99UN, HspE7, FP253/Fludarabine, ALVAC(2) melanoma multi-antigen therapeutic vaccine, ALVAC-hB7.1, canarypox-hIL-12 melanoma vaccine, Ad-REIC/Dkk-3, rAd-IFN SCH 721015, TIL-Ad-INFg, Ad-ISF35, and coxsackievirus A21 (CVA21, CAVATAK®).

In other embodiments, anti-TCRβV antibody molecule, multispecific or multifunctional molecule is administered in combination with a nanopharmaceutical. Exemplary cancer nanopharmaceuticals include, but not limited to, ABRAXANE® (paclitaxel bound albumin nanoparticles), CRLX101 (CPT conjugated to a linear cyclodextrin-based polymer), CRLX288 (conjugating docetaxel to the biodegradable polymer poly (lactic-co-glycolic acid)), cytarabine liposomal (liposomal Ara-C, DEPOCYT™), daunorubicin liposomal (DAUNOXOME®), doxorubicin liposomal (DOXIL®, CAELYX®), encapsulated-daunorubicin citrate liposome (DAUNOXOME®), and PEG anti-VEGF aptamer (MACUGEN®).

In some embodiments, the anti-TCRβV antibody molecule, multispecific or multifunctional molecule is administered in combination with paclitaxel or a paclitaxel formulation, e.g., TAXOL®, protein-bound paclitaxel (e.g., ABRAXANE®). Exemplary paclitaxel formulations include, but are not limited to, nanoparticle albumin-bound paclitaxel (ABRAXANE®, marketed by Abraxis Bioscience), docosahexaenoic acid bound-paclitaxel (DHA-paclitaxel, Taxoprexin, marketed by Protarga), polyglutamate bound-paclitaxel (PG-paclitaxel, paclitaxel poliglumex, CT-2103, XYOTAX, marketed by Cell Therapeutic), the tumor-activated prodrug (TAP), ANG105 (Angiopep-2 bound to three molecules of paclitaxel, marketed by ImmunoGen), paclitaxel-EC-1 (paclitaxel bound to the erbB2-recognizing peptide EC-1; see Li et al., Biopolymers (2007) 87:225-230), and glucose-conjugated paclitaxel (e.g., 2′-paclitaxel methyl 2-glucopyranosyl succinate, see Liu et al., Bioorganic & Medicinal Chemistry Letters (2007) 17:617-620).

Exemplary RNAi and antisense RNA agents for treating cancer include, but not limited to, CALAA-01, siG12D LODER (Local Drug EluteR), and ALN-VSP02.

Other cancer therapeutic agents include, but not limited to, cytokines (e.g., aldesleukin (IL-2, Interleukin-2, PROLEUKIN®), alpha Interferon (IFN-alpha, Interferon alfa, INTRON® A (Interferon alfa-2b), ROFERON-A® (Interferon alfa-2a)), Epoetin alfa (PROCRIT®), filgrastim (G-CSF, Granulocyte—Colony Stimulating Factor, NEUPOGEN®), GM-CSF (Granulocyte Macrophage Colony Stimulating Factor, sargramostim, LEUKINE™), IL-11 (Interleukin-11, oprelvekin, NEUMEGA®), Interferon alfa-2b (PEG conjugate) (PEG interferon, PEG-INTRON™), and pegfilgrastim (NEULASTA™)), hormone therapy agents (e.g., aminoglutethimide (CYTADREN®), anastrozole (ARIMIDEX®), bicalutamide (CASODEX®), exemestane (AROMASIN®), fluoxymesterone (HALOTESTIN®), flutamide (EULEXIN®), fulvestrant (FASLODEX®), goserelin (ZOLADEX®), letrozole (FEMARA®), leuprolide (ELIGARD™, LUPRON®, LUPRON DEPOT®, VIADUR™), megestrol (megestrol acetate, MEGACE®), nilutamide (ANANDRON®, NILANDRON®), octreotide (octreotide acetate, SANDOSTATIN®, SANDOSTATIN LAR®), raloxifene (EVISTA®), romiplostim (NPLATE®), tamoxifen (NOVALDEX®), and toremifene (FARESTON®)), phospholipase A2 inhibitors (e.g., anagrelide (AGRYLIN®)), biologic response modifiers (e.g., BCG (THERACYS®, TICE®), and Darbepoetin alfa (ARANESP®)), target therapy agents (e.g., bortezomib (VELCADE®), dasatinib (SPRYCEL™), denileukin diftitox (ONTAK®), erlotinib (TARCEVA®), everolimus (AFINITOR®), gefitinib (IRESSA®), imatinib mesylate (STI-571, GLEEVEC™), lapatinib (TYKERB®), sorafenib (NEXAVAR®), and SU11248 (sunitinib, SUTENT®)), immunomodulatory and antiangiogenic agents (e.g., CC-5013 (lenalidomide, REVLIMID®), and thalidomide (THALOMID®)), glucocorticosteroids (e.g., cortisone (hydrocortisone, hydrocortisone sodium phosphate, hydrocortisone sodium succinate, ALA-CORT®, HYDROCORT ACETATE®, hydrocortone phosphate LANACORT®, SOLU-CORTEF®), decadron (dexamethasone, dexamethasone acetate, dexamethasone sodium phosphate, DEXASONE®, DIODEX®, HEXADROL®, MAXIDEX®), methylprednisolone (6-methylprednisolone, methylprednisolone acetate, methylprednisolone sodium succinate, DURALONE®, MEDRALONE®, MEDROL®, M-PREDNISOL®, SOLU-MEDROL®), prednisolone (DELTA-CORTEF®, ORAPRED®, PEDIAPRED®, PRELONE®), and prednisone (DELTASONE®, LIQUID PRED®, METICORTEN®, ORASONE®)), and bisphosphonates (e.g., pamidronate (AREDIA®), and zoledronic acid (ZOMETA®))

In some embodiments, the anti-TCRβV antibody molecule, multispecific or multifunctional molecule is used in combination with a tyrosine kinase inhibitor (e.g., a receptor tyrosine kinase (RTK) inhibitor). Exemplary tyrosine kinase inhibitor include, but are not limited to, an epidermal growth factor (EGF) pathway inhibitor (e.g., an epidermal growth factor receptor (EGFR) inhibitor), a vascular endothelial growth factor (VEGF) pathway inhibitor (e.g., an antibody against VEGF, a VEGF trap, a vascular endothelial growth factor receptor (VEGFR) inhibitor (e.g., a VEGFR-1 inhibitor, a VEGFR-2 inhibitor, a VEGFR-3 inhibitor)), a platelet derived growth factor (PDGF) pathway inhibitor (e.g., a platelet derived growth factor receptor (PDGFR) inhibitor (e.g., a PDGFR-β inhibitor)), a RAF-1 inhibitor, a KIT inhibitor and a RET inhibitor. In some embodiments, the anti-cancer agent used in combination with the AHCM agent is selected from the group consisting of: axitinib (AG013736), bosutinib (SKI-606), cediranib (RECENTIN™, AZD2171), dasatinib (SPRYCEL®, BMS-354825), erlotinib (TARCEVA®), gefitinib (IRESSA®), imatinib (Gleevec®, CGP57148B, STI-571), lapatinib (TYKERB®, TYVERB®), lestaurtinib (CEP-701), neratinib (HKI-272), nilotinib (TASIGNA®), semaxanib (semaxinib, SU5416), sunitinib (SUTENT®, SU11248), toceranib (PALLADIA®), vandetanib (ZACTIMA®, ZD6474), vatalanib (PTK787, PTK/ZK), trastuzumab (HERCEPTIN®), bevacizumab (AVASTIN®), rituximab (RITUXAN®), cetuximab (ERBITUX®), panitumumab (VECTIBIX®), ranibizumab (Lucentis®), nilotinib (TASIGNA®), sorafenib (NEXAVAR®), alemtuzumab (CAMPATH®), gemtuzumab ozogamicin (MYLOTARG®), ENMD-2076, PCI-32765, AC220, dovitinib lactate (TKI258, CHIR-258), BMW 2992 (TOVOK™), SGX523, PF-04217903, PF-02341066, PF-299804, BMS-777607, ABT-869, MP470, BIBF 1120 (VARGATEF®), AP24534, JNJ-26483327, MGCD265, DCC-2036, BMS-690154, CEP-11981, tivozanib (AV-951), OSI-930, MM-121, XL-184, XL-647, XL228, AEE788, AG-490, AST-6, BMS-599626, CUDC-101, PD153035, pelitinib (EKB-569), vandetanib (zactima), WZ3146, WZ4002, WZ8040, ABT-869 (linifanib), AEE788, AP24534 (ponatinib), AV-951 (tivozanib), axitinib, BAY 73-4506 (regorafenib), brivanib alaninate (BMS-582664), brivanib (BMS-540215), cediranib (AZD2171), CHIR-258 (dovitinib), CP 673451, CYC116, E7080, Ki8751, masitinib (AB1010), MGCD-265, motesanib diphosphate (AMG-706), MP-470, OSI-930, Pazopanib Hydrochloride, PD173074, Sorafenib Tosylate (Bay 43-9006), SU 5402, TSU-68 (SU6668), vatalanib, XL880 (GSK1363089, EXEL-2880). Selected tyrosine kinase inhibitors are chosen from sunitinib, erlotinib, gefitinib, or sorafenib. In one embodiment, the tyrosine kinase inhibitor is sunitinib.

In one embodiment, the anti-TCRβV antibody molecule, multispecific or multifunctional molecule is administered in combination with one of more of: an anti-angiogenic agent, or a vascular targeting agent or a vascular disrupting agent. Exemplary anti-angiogenic agents include, but are not limited to, VEGF inhibitors (e.g., anti-VEGF antibodies (e.g., bevacizumab); VEGF receptor inhibitors (e.g., itraconazole); inhibitors of cell proliferatin and/or migration of endothelial cells (e.g., carboxyamidotriazole, TNP-470); inhibitors of angiogenesis stimulators (e.g., suramin), among others. A vascular-targeting agent (VTA) or vascular disrupting agent (VDA) is designed to damage the vasculature (blood vessels) of cancer tumors causing central necrosis (reviewed in, e.g., Thorpe, P.E. (2004) Clin. Cancer Res. Vol. 10:415-427). VTAs can be small-molecule. Exemplary small-molecule VTAs include, but are not limited to, microtubule destabilizing drugs (e.g., combretastatin A-4 disodium phosphate (CA4P), ZD6126, AVE8062, Oxi 4503); and vadimezan (ASA404).

Immune Checkpoint Inhibitors

In other embodiments, methods described herein comprise use of an immune checkpoint inhibitor in combination with the anti-TCRβV antibody molecule, multispecific or multifunctional molecule. The methods can be used in a therapeutic protocol in vivo.

In embodiments, an immune checkpoint inhibitor inhibits a checkpoint molecule. Exemplary checkpoint molecules include but are not limited to CTLA4, PD1, PD-L1, PD-L2, LAG3, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), BTLA, KIR, MHC class I, MHC class II, GALS, VISTA, BTLA, TIGIT, LAIR1, and A2aR. See, e.g., Pardoll. Nat. Rev. Cancer 12.4(2012):252-64, incorporated herein by reference.

In embodiments, the immune checkpoint inhibitor is a PD-1 inhibitor, e.g., an anti-PD-1 antibody such as Nivolumab, Pembrolizumab or Pidilizumab. Nivolumab (also called MDX-1106, MDX-1106-04, ONO-4538, or BMS-936558) is a fully human IgG4 monoclonal antibody that specifically inhibits PD1. See, e.g., U.S. Pat. No. 8,008,449 and WO2006/121168. Pembrolizumab (also called Lambrolizumab, MK-3475, MK03475, SCH-900475 or KEYTRUDA®; Merck) is a humanized IgG4 monoclonal antibody that binds to PD-1. See, e.g., Hamid, O. et al. (2013) New England Journal of Medicine 369 (2): 134-44, U.S. Pat. No. 8,354,509 and WO2009/114335. Pidilizumab (also called CT-011 or Cure Tech) is a humanized IgG1k monoclonal antibody that binds to PD1. See, e.g., WO2009/101611. In one embodiment, the inhibitor of PD-1 is an antibody molecule having a sequence substantially identical or similar thereto, e.g., a sequence at least 85%, 90%, 95% identical or higher to the sequence of Nivolumab, Pembrolizumab or Pidilizumab. Additional anti-PD1 antibodies, e.g., AMP 514 (Amplimmune), are described, e.g., in U.S. Pat. No. 8,609,089, US 2010028330, and/or US 20120114649.

In some embodiments, the PD-1 inhibitor is an immunoadhesin, e.g., an immunoadhesin comprising an extracellular/PD-1 binding portion of a PD-1 ligand (e.g., PD-L1 or PD-L2) that is fused to a constant region (e.g., an Fc region of an immunoglobulin). In embodiments, the PD-1 inhibitor is AMP-224 (B7-DCIg, e.g., described in WO2011/066342 and WO2010/027827), a PD-L2 Fc fusion soluble receptor that blocks the interaction between B7-H1 and PD-1.

In embodiments, the immune checkpoint inhibitor is a PD-L1 inhibitor, e.g., an antibody molecule. In some embodiments, the PD-L1 inhibitor is YW243.55.S70, MPDL3280A, MEDI-4736, MSB-0010718C, or MDX-1105. In some embodiments, the anti-PD-L1 antibody is MSB0010718C (also called A09-246-2; Merck Serono), which is a monoclonal antibody that binds to PD-L1. Exemplary humanized anti-PD-L1 antibodies are described, e.g., in WO2013/079174. In one embodiment, the PD-L1 inhibitor is an anti-PD-L1 antibody, e.g., YW243.55.S70. The YW243.55.S70 antibody is described, e.g., in WO 2010/077634. In one embodiment, the PD-L1 inhibitor is MDX-1105 (also called BMS-936559), which is described, e.g., in WO2007/005874. In one embodiment, the PD-L1 inhibitor is MDPL3280A (Genentech/Roche), which is a human Fc-optimized IgG1 monoclonal antibody against PD-L1. See, e.g., U.S. Pat. No. 7,943,743 and U.S. Publication No.: 20120039906. In one embodiment, the inhibitor of PD-L1 is an antibody molecule having a sequence substantially identical or similar thereto, e.g., a sequence at least 85%, 90%, 95% identical or higher to the sequence of YW243.55.570, MPDL3280A, MEDI-4736, MSB-0010718C, or MDX-1105.

In embodiments, the immune checkpoint inhibitor is a PD-L2 inhibitor, e.g., AMP-224 (which is a PD-L2 Fc fusion soluble receptor that blocks the interaction between PD1 and B7-H1. See, e.g., WO2010/027827 and WO2011/066342.

In one embodiment, the immune checkpoint inhibitor is a LAG-3 inhibitor, e.g., an anti LAG-3 antibody molecule. In embodiments, the anti-LAG-3 antibody is BMS-986016 (also called BMS986016; Bristol-Myers Squibb). BMS-986016 and other humanized anti-LAG-3 antibodies are described, e.g., in US 2011/0150892, WO2010/019570, and WO2014/008218.

In embodiments, the immune checkpoint inhibitor is a TIM-3 inhibitor, e.g., anti-TIM3 antibody molecule, e.g., described in U.S. Pat. No. 8,552,156, WO 2011/155607, EP 2581113 and U.S. Publication No.: 2014/044728.

In embodiments, the immune checkpoint inhibitor is a CTLA-4 inhibitor, e.g., anti-CTLA-4 antibody molecule. Exemplary anti-CTLA4 antibodies include Tremelimumab (IgG2 monoclonal antibody from Pfizer, formerly known as ticilimumab, CP-675,206); and Ipilimumab (also called MDX-010, CAS No. 477202-00-9). Other exemplary anti-CTLA-4 antibodies are described, e.g., in U.S. Pat. No. 5,811,097.

CRS Grading

In some embodiments, CRS can be graded in severity from 1-5 as follows. Grades 1-3 are less than severe CRS. Grades 4-5 are severe CRS. For Grade 1 CRS, only symptomatic treatment is needed (e.g., nausea, fever, fatigue, myalgias, malaise, headache) and symptoms are not life threatening. For Grade 2 CRS, the symptoms require moderate intervention and generally respond to moderate intervention. Subjects having Grade 2 CRS develop hypotension that is responsive to either fluids or one low-dose vasopressor; or they develop grade 2 organ toxicity or mild respiratory symptoms that are responsive to low flow oxygen (<40% oxygen). In Grade 3 CRS subjects, hypotension generally cannot be reversed by fluid therapy or one low-dose vasopressor. These subjects generally require more than low flow oxygen and have grade 3 organ toxicity (e.g., renal or cardiac dysfunction or coagulopathy) and/or grade 4 transaminitis. Grade 3 CRS subjects require more aggressive intervention, e.g., oxygen of 40% or higher, high dose vasopressor(s), and/or multiple vasopressors. Grade 4 CRS subjects suffer from immediately life-threatening symptoms, including grade 4 organ toxicity or a need for mechanical ventilation. Grade 4 CRS subjects generally do not have transaminitis. In Grade 5 CRS subjects, the toxicity causes death. Sets of criteria for grading CRS are provided herein as Table 5, Table 6, and Table 7. Unless otherwise specified, CRS as used herein refers to CRS according to the criteria of Table 6.

In embodiments, CRS is graded according to Table 5:

TABLE 5 CRS grading Gr1 Supportive care only Gr2 IV therapies +/− hospitalization. Gr3 Hypotension requiring IV fluids or low-dose vasoactives or hypoxemia requiring oxygen, CPAP, or BIPAP. Gr4 Hypotension requiring high-dose vasoactives or hypoxemia requiring mechanical ventilation. Gr 5 Death

TABLE 6 CTCAE v 4.0 CRS grading scale CRS grade Characteristics Grade 1 Mild; No infusion interruption; No intervention Grade 2 Infusion interruption indicated but responds promptly to symptomatic treatment (e.g., antihistamines, NSAIDS, narcotics, IV fluids); prophylactic medications indicated for <=24 hrs Grade 3 Prolonged (e.g., not rapidly responsive to symptomatic medications and/or brief interruption of infusion); recurrence of symptoms following initial improvement; hospitalization indicated for clinical sequelae (e.g., renal impairment, pulmonary infiltrates) Grade 4 Life threatening consequences; pressor or ventilator support

TABLE 7 NCI CRS grading scale CRS grade Characteristics Grade 1 Symptoms are not life threatening and require symptomatic treatment only; e.g., fever, nausea, fatigue, headache, myalgias, malaise Grade 2 Symptoms require and respond to moderate intervention; Oxygen requirement <40% or hypotension responsive to fluids or low dose pressors or Grade 2 organ toxicity Grade 3 Symptoms require and respond to aggressive intervention; Oxygen requirement >=40% or Hypotension requiring high dose or multiple pressors or grade 3 organ toxicity or grade 4 transaminitis Grade 4 Life threatening symptoms Requirement for ventilator support or Grade 4; organ toxicity (excluding transaminitis)

EXAMPLES Example 1. Humanization of α-TRBV6-5 Antibody Clone Antibody A

The germline for the mouse α-TCRβ antibody clone Antibody A VH and VL were assigned using IMGT nomenclature, with CDR regions defined by a combined Kabat and Chothia classification. SEQ ID NO: 1 and SEQ ID NO: 2 are the Antibody A VH and VL sequences respectively where the VH germline is mouse IGHV1S12*01 and the VL germline is mouse IGKV6-15*01. SEQ ID NOs: 3-5 are the Antibody A VH CDR regions 1-3 respectively and SEQ ID NOs: 6-8 correspond to the VL CDR regions 1-3 (as described in Table 1).

Humanization of the Antibody A VH and VL sequences was done separately using similar methodology. Amino acids positions were identified in the framework regions which were important for the success of CDR grafting. Human germline sequences were identified which preserved the necessary residues and contained a high amount of overall identity. When the human germline framework sequence did not contain a matching important amino acid, it was back mutated to match the mouse sequence. CDR regions were grafted onto the human germline unchanged. The Antibody A VH was humanized into human IGHV1-69*01 and the Antibody A VL was humanized into IGKV1-17*01 and IGKV1-27*01. All 3 humanized sequences were confirmed to contain no introduced potential negative post translational modification sites such as NG, DG, NS, NN, DS, NT, NXS, or NXT as a result of the humanization process. SEQ ID NO: 9 is the humanized Antibody A-H.1 VH and SEQ ID NOs: 10 and 11 are the humanized VL IGKV1-17*01 and IGKV1-27*01 germlines respectively (as described in Table 1). FIGS. 1A and 1B show the murine and humanized sequences with annotations depicting the CDR and framework regions (FR).

Example 2: Humanization of α-TRBV12-3 and TRBV12-4 Antibody Clone Antibody B

The germline for the mouse α-TCRβ antibody clone Antibody B VH and VL were assigned using IMGT nomenclature, with CDR regions defined by a combined Kabat and Chothia classification. SEQ ID NO: 15 and SEQ ID NO: 16 are the Antibody B VH and VL sequences respectively where the VH germline is mouse IGHV5-17*02 and the VL germline is mouse IGKV4-50*01. SEQ ID NOs: 17-19 are the B-H VH CDR regions 1-3 respectively and SEQ ID NOs: 20-22 are the B-H VL CDR regions 1-3 (as described in Table 2).

The method applied to humanize Antibody A described in Example 1 was used to humanize Antibody B. The Antibody B VH was humanized into human IGHV3-30*01, IGHV3-48*01, and IGHV3-66*01 and the Antibody B VL was humanized into human IGKV1-9*01, IGKV1-39*01, IGKV3-15*01, IGLV1-47*01 and IGLV3-10*01. SEQ ID NOs: 23-25 are the B-H.1A, B-H.1B, and B-H.1C humanized heavy chains and SEQ ID NOs: 26-30 are the B-H.1D, B-H.1E, B-H.1F, B-H.1G and B-H.1H humanized light chains (as described in Table 2). FIGS. 2A and 2B show the murine and humanized sequences with annotations depicting the CDR and framework regions (FR).

Example 3: Characteristics of Anti-TCRβV Antibodies

Introduction

Current bispecific constructs designed to redirect T cells to promote tumor cell lysis for cancer immunotherapy typically utilize single chain variable fragments (scFvs) that are derived from monoclonal antibodies (mAb) directed against the CD3e subunit of the T cell receptor (TCR). However, there are limitations to this approach which may prevent the full realization of the therapeutic potential for such bispecific constructs. Previous studies have shown that, e.g., low “activating” doses of anti-CD3e mAb can cause long-term T cell dysfunction and exert immunosuppressive effects. In addition, anti-CD3e mAbs bind to all T cells and thus activate equally all T cells, which has been associated with the first dose side effects of anti-CD3e mAbs that result from massive T cell activation. These large number of activated T cells secrete substantial amounts of cytokines, the most important of which is Interferon gamma (IFNg). This excess amount of IFNg in turn, e.g., activates macrophages which then can overproduce proinflammatory cytokines such as IL-1, IL-6 and TNF-alpha, causing a “cytokine storm” known as the cytokine release syndrome (CRS). Thus, it might be advantageous to develop antibodies that are capable of binding and activating only a subset of necessary effector T cells to reduce the CRS.

Results

To that end, antibodies directed to the variable chain of the beta subunit of TCR (TCR Vb) were identified. These anti-TCR Vb antibodies bind and activate a subset of T cells, but with, e.g., no or markedly reduced CRS. Using plate-bound anti-TCR Vb13.1 mAbs (A-H.1 and A-H.2) it was shown that a population of T cells, defined by positive staining with A-H.1, can be expanded (from ˜5% of T cells on day 0 to almost 60% of total T cells on day 6 of cell culture) (FIGS. 4A-4C). For this experiment, human CD3+ T cells were isolated using magnetic-bead separation (negative selection) and activated with immobilized (plate-coated) A-H.1 or OKT3 (anti-CD3e) antibodies at 100 nM for 6 days. The expanded Vb13.1+ T cells display cytolytic activity against transformed cell line RPMI-8226 when co-cultured with purified CD3+ T cells (FIGS. 5A-5B).

Next, the ability of PBMCs activated by anti-TCR VB antibodies to produce cytokines was assessed. The cytokine production of PBMCs activated with anti-TCR VB antibodies was compared to the cytokine production of PBMCs activated with: (i) anti-CD3e antibodies (OKT3 or SP34-2); (ii) anti-TCR V alpha (TCR VA) antibodies including anti-TCR VA 12.1 antibody 6D6.6, anti-TCR VA24JA18 antibody 6B11; (iii) anti-TCR alpha beta antibody T10B9; and/or (iv) isotype control (BGM0109). The anti-TCR VB antibodies tested include: humanized anti-TCRVB 13.1 antibodies (A-H.1, or A-H.2), murine anti-TCR VB5 antibody Antibody E, murine anti-TCR VB8.1 antibody Antibody B, and murine anti-TCR VB12 antibody Antibody D. BGM0109 comprises the amino acid sequence of

(SEQ ID NO: 3282) METDTLLLWVLLLWVPGSTGGLNDIFEAQKIEWHEGGGGSEPRTDTDTCP NPPDPCPTCPTPDLLGGPSVFIFPPKPKDVLMISLTPKITCVVVDVSEEE PDVQFNWYVNNVEDKTAQTETRQRQYNSTYRVVSVLPIKHQDWMSGKVFK CKVNNNALPSPIEKTISKPRGQVRVPQIYTFPPPIEQTVKKDVSVTCLVT GFLPQDIHVEWESNGQPQPEQNYKNTQPVLDSDGSYFLYSKLNVPKSRWD QGDSFTCSVIHEALHNHHMTKTISRSLGNGGGGS.

As shown in FIG. 6A, when plate-bound A-H.1 or A-H.2, or anti-CD3e antibodies (OKT3 or SP34-2) were used to activate human PBMCs, the T cell cytokine IFNg was induced (FIG. 6A). All anti-TCR VB antibodies tested had a similar effect on the production of IFNg (FIG. 6B). The anti-TCR VA antibodies did not induce similar IFNg production.

With respect to IL-2 production, PBMCs activated with A-H.1 and A-H.2 resulted in increased IL-2 production (FIG. 7A) with delayed kinetics (FIG. 7B) as compared to PBMCs activated with anti-CD3e antibodies (OKT3 or SP34-2). FIG. 7B shows that anti-TCR VB antibody activated PBMCs demonstrate peak production of IL-2 at Day 5 or Day 6 post-activation (incubation with plate-coated antibodies). In contrast, IL-2 production in PBMCs activated with OKT3 peaked at day 2 post-activation. As with IFNG, the IL-2 effect (e.g., enhanced production of IL-2 and delayed kinetics) was similar across all anti-TCR VB antibodies tested (FIG. 7B).

The production of cytokines IL-6, IL-1β and TNF-alpha which are associated with “cytokine storms” (and accordingly CRS) was also assessed under similar conditions. FIGS. 8A, 9A and 10A shows that while PBMCs activated with anti-CD3e antibodies demonstrate production of IL-6 (FIG. 8A), TNF-alpha (FIG. 9A) and IL-1β (FIG. 10A), no or little induction of these cytokines was observed with PBMCs activated with A-H.1 or A-H.2. As shown in FIGS. 9B and 10B, TNF-alpha and IL-1β production was not induced by activation of PBMCs with any of the anti-TCR VB antibodies.

It was further noted that the kinetics of IFNg production by A-H.1-activated CD3+ T cells was delayed relative to those produced by CD3+ T cells activated by anti-CD3e mAbs (OKT3 and SP34-2) (FIGS. 11A and 11B).

Finally, it was observed that the subset of memory effector T cells known as TEMRA was preferentially expanded in CD8+ T cells activated by A-H.1 or A-H.2 (FIG. 12). Isolated human PBMCs were activated with immobilized (plate-coated) anti-CD3e or anti-TCR Vβ13.1 at 100 nM for 6-days. After a 6-day incubation, T-cell subsets were identified by FACS staining for surface markers for Naïve T cell (CD8+, CD95−, CD45RA+, CCR7+), T stem cell memory (TSCM; CD8+, CD95+, CD45RA+, CCR7+), T central memory (Tcm; CD8+, CD95+, CD45RA−, CCR7+), T effector memory (Tem; CD8+, CD95+, CD45RA−, CCR7−), and T effector memory re-expressing CD45RA (Temra; CD8+, CD95+, CD45RA+, CCR7−). Human PBMCs activated by anti-TCR Vβ13.1 antibodies (A-H.1 or A-H.2) increased CD8+ TSCM and Temra T cell subsets when compared to PBMCs activated by anti-CD3e antibodies (OKT3 or SP34-2). Similar expansion was observed with CD4+ T cells.

Conclusion

The data provided in this Example show that antibodies directed against TCR Vb can, e.g., preferentially activate a subset of T cells, leading to an expansion of TEMRA, which can, e.g., promote tumor cell lysis but not CRS. Thus, bispecific constructs utilizing either a Fab or scFV or a peptide directed to the TCR Vb can, e.g., be used to activate and redirect T cells to promote tumor cell lysis for cancer immunotherapy, without, e.g., the harmful side-effects of CRS associated with anti-CD3e targeting.

Example 4: On-Target T Cell Mediated Cytotoxicity of Multiple Myeloma (MM) Cells with a Dual-Targeting Antibody Molecule Against BCMA and a T Cell Engager

This example shows on-target T cell mediated cytotoxicity of multiple myeloma (MM) cells with dual-targeting antibody molecules that recognize a T cell engager, e.g., TCRVb, on T cells and BCMA on MM cells.

As shown in FIG. 13A, purified human T cells activated with plate-bound anti-TCRVb antibody for 5 days proliferate at a higher rate than purified human T cells activated with plate-bound anti-CD3 (OKT3) antibody. Anti-TCRVb antibody stimulation of T cells resulted in selective expansion of CD45RA+ effector memory CD8+ and CD4+ T cells (TEMRA) cells (FIG. 13B). Both CD8+ and CD4+ Temra cell populations expanded more when stimulated with an anti-TCRVb antibody, compared to unstimulated cells or cells stimulated with an anti-CD3 (SP34) antibody. Anti-TCRVb antibodies resulted in delayed secretion of IFN-g by PBMCs stimulated with an anti-TCRVb antibody compared to PBMCs stimulated with anti-CD3 antibodies (FIG. 13C). Additionally, T cells stimulated with anti-TCRVb antibody or anti-CD3 antibodies resulted in comparable lysis of multiple myeloma target cells, as shown in FIG. 13D. As shown in FIGS. 13E-13F, T cells stimulated for 5 days with 100 ng/ml plate-bound an anti-TCRVb antibody, or an anti-CD3 antibody secreted perforin and Granzyme B.

Activation of PBMCs with anti-TCRVb antibody resulted in higher production and/or secretion of IL-2 and/or IL-15 compared to PBMCs activated with an anti-OKT3 antibody (FIG. 14A). Anti-TCRVb antibody activated of PBMCs also resulted in expansion and/or survival, e.g., proliferation of Natural Killer (NK) cells (FIG. 14B). In comparison, PBMCS activated with an anti-OKT3 antibody did not result in NK cell expansion. Further, as described in Example 3, PBMCs activated with an anti-TCRVb antibody did not result in the production of cytokines IL-6, IL-1β and TNF-alpha which are associated with CRS (FIG. 15). These in vitro characterization studies show that in some embodiments, anti-TCRVb antibodies, e.g., activate and/or stimulate, T cells to promote T cell killing as evidenced by target cell lysis, perforin secretion and granzyme B secretion, and secretion of IFN-g with, e.g., delayed kinetics.

Next, the ability of a dual-targeting antibody molecule (Molecule I), which targets BCMA on one arm and TCRVb on the other arm, to target and kill multiple myeloma (MM) cells was tested. Healthy donor PBMCs were co-incubated with the RMPI8226 MM cell line and one of the following dual-targeting antibody molecules: BCMA-TCRVb (Molecule I), BCMA-CD3, or Control-TCRVb; or an isotype control Target cell lysis was then assessed using flow cytometry. As shown in FIG. 16A, the dual-targeting BCMA-TCRVb antibody molecule (Molecule I) resulted in killing of MM cells in vitro.

The dual-targeting BCMA-TCRVb antibody molecule (Molecule I) was further tested in vivo for its ability to inhibit MM tumor growth in a MM mouse model. The NCI-H929 cell line was injected in NOD-scid IL2rγnull (NSG) recipient mice on Day 0 followed by delivery of PBMCs on Day 9. On Days 12, 15, 18 and 21, the dual-targeting BCMA-TCRVb antibody molecule (Molecule I) was administered via intraperitoneal injection at a dose of 0.5 mg/kg. FIG. 16B shows prevention, e.g., inhibition, of MM tumor growth in vivo with the dual-targeting BCMA-TCRVb antibody molecule (Molecule I). These results demonstrate that in some embodiments the dual-targeting BCMA-TCRVb antibody molecule, e.g., can kill tumor cells, e.g., MM tumor cells, in vitro and in vivo. Accordingly, in some embodiments, a dual-targeting BCMA-TCRVb antibody molecule can be used, e.g., as a therapy for cancer, e.g., a hematological cancer, e.g., MM.

Example 5: In Vitro Cytotoxicity of a Dual-Targeting Antibody Molecule Against FcRH5 and a T Cell Engager

This example shows in vitro cytotoxicity on multiple myeloma (MM) cells with a dual-targeting antibody molecule that recognizes a T cell engager, e.g., TCRVb, on T cells and FcRH5 on MM cells. Healthy donor PBMCs or purified T cells were co-incubated with the MOL8M MM cell line and a dual-targeting antibody molecule which targets FcRH5 on one arm and TCRVb on the other arm (Molecule E), or with an isotype control antibody. Target cell lysis was then assessed using flow cytometry. As shown in FIG. 17, the dual targeting FcRH5-TCRVb molecule (Molecule E) resulted in killing of MM cells by both purified T cells or PBMCs. This shows that the dual targeting FcRH5-TCRVb molecule can target and promote killing of MM cells by immune cells, e.g., in PBMCs, including T cells.

Example 6: Characteristics of Anti-TCR Vβ8a Antibodies

This Example shows in vitro characterization of anti-TCR Vβ8a antibodies (B-H.1). TCR V138 is also referred to as TCR V1312 (as described in Table 8A). Isolated human PBMCs were activated with immobilized (plate-coated) anti-CD3E or anti-TCR Vβ8a at 100 nM, and cell culture supernatants were collected on day 1, 2, 3, 5, 6 and 8 post stimulation. Cytokines (IFNγ, IL-2, TNFα, IL-1β or IL-6) were measured using MSD technology platform (MesoScale Discovery) as described in the manufacturer's protocol.

As shown in FIGS. 18A-18B, Human PBMCs activated by anti-TCR Vβ8a antibodies (B-H.1) produce similar or reduced levels of IFNγ (FIG. 18A) and higher levels IL-2 (FIG. 18B) when compared to those activated by anti-CD3E antibodies (OKT3 or SP34-2).

FIGS. 19A-19B show that human PBMCs activated by anti-TCR Vβ8a antibodies (B-H.1) do not produce significant levels of IL-6, or IL1b. Activation of human PBMCs with anti-TCR Vβ8a antibodies (B-H.1) also results in lesser TNFα when compared to PBMCs activated by anti-CDR antibodies (OKT3 or SP34-2) (see FIG. 19C).

In summary, as shown in Example 3, this Example shows that anti-TCR Vβ8a antibodies can, e.g., preferentially induce expression of T cell cytokines, e.g., IL-2 and IFNg, but not production of cytokines IL-6, IL-1β and TNF-alpha which are associated with “cytokine storms” (and accordingly CRS).

Example 7: Characteristics of Anti-TCRβV Antibody D Antibody

This Example describes characterization of anti-TCRβV antibodies which can bind and activate a subset of T cells, but with, e.g., no or markedly reduced, CRS.

Human PBMCs were isolated from whole blood followed by solid-phase (plate-coated) stimulation with anti-TCR Vβ12 antibody (Antibody D) or anti-CD3e antibodies (OKT3) at 100 nM. Supernatant was collected on Days 1, 2, 3, 5, or 6 followed by multiplex cytokine analysis for IFNg, IL-2, IL-6, IL-1beta, or TNFalpha. The data was quantified using MSD (Meso Scale Discovery) platform, following the manufacturer's protocol.

As shown in FIG. 20A, when plate-bound anti-TCR Vβ12 antibody (Antibody D) or anti-CD3e antibodies (OKT3) were used to activate human PBMCs, the T cell cytokine IFNg was induced. With respect to IL-2 production, PBMCs activated with anti-TCR Vβ12 antibody (Antibody D) resulted in increased IL-2 production with delayed kinetics (FIG. 20B) as compared to PBMCs activated with anti-CD3e antibodies (OKT3).

The production of cytokines IL-6, IL-10 and TNF-alpha which are associated with “cytokine storms” (and accordingly CRS) was also assessed under similar conditions. FIGS. 20C-20E show that that while PBMCs activated with anti-CD3e antibodies demonstrate production of IL-6 (FIG. 20D), TNF-alpha (FIG. 20C) and IL-1β (FIG. 20E), no or little induction of these cytokines was observed with PBMCs activated with anti-TCR Vβ12 antibody (Antibody D).

The data provided in this Example show that antibodies directed against TCR VP can, e.g., preferentially activate a subset of T cells, and do not results in induction of cytokines associated with cytokine storms or CRS.

Example 8: Characteristics of Anti-TCRβV Antibody E

This Example describes characterization of anti-TCRβV antibodies which can bind and activate a subset of T cells, but with, e.g., no or markedly reduced, CRS.

Human PBMCs were isolated from whole blood followed by solid-phase (plate-coated) stimulation with anti-TCR Vβ5 antibody (Antibody E) or anti-CD3e antibodies (OKT3 and SP34-2), each at 100 nM. Supernatant was collected on Days 1, 3, 5, or 7 followed by multiplex cytokine analysis for IFNg, IL-2, IL-6, IL-1beta, IL-10 or TNFalpha. The data was quantified using MSD (Meso Scale Discovery) platform, following the manufacturer's protocol.

As shown in FIG. 21A, when plate-bound anti-TCR Vβ5 antibody (Antibody E) or anti-CD3e antibodies (OKT3 and SP34-2) were used to activate human PBMCs, the T cell cytokine IFNg was induced. With respect to IL-2 production, PBMCs activated with anti-TCR Vβ5 antibody (Antibody E) resulted in increased IL-2 production with delayed kinetics (FIG. 21B) as compared to PBMCs activated with anti-CD3e antibodies (OKT3 or SP34-2).

The production of cytokines IL-6, IL-1β, IL-10 and TNF-alpha which are associated with “cytokine storms” (and accordingly CRS) was also assessed under similar conditions. FIGS. 22A-22D show that that while PBMCs activated with anti-CD3e antibodies demonstrate production of IL-1β (FIG. 22A), IL-6 (FIG. 22B), TNF-alpha (FIG. 22C) and IL-10 (FIG. 22D), no or little induction of these cytokines was observed with PBMCs activated with anti-TCR Vβ5 antibody (Antibody E).

The data provided in this Example show that antibodies directed against TCR VP can, e.g., preferentially activate a subset of T cells, and do not results in induction of cytokines associated with cytokine storms or CRS.

Example 9: Characteristics of a Dual-Targeting Antibody Molecule Against BCMA and TCRβV

This Example describes characterization of a dual targeting antibody (e.g., a bispecific molecule) comprising an anti-TCRβV binding moiety and a BCMA binding moiety (Molecule H) which can bind and activate a subset of T cells, but with, e.g., no or markedly reduced, CRS.

Human PBMCs were isolated from whole blood followed by solid-phase (plate-coated) stimulation with an anti-TCRβV x BCMA bispecific molecule (Molecule H) or anti-CD3e antibodies (OKT3), each at 100 nM. Supernatant was collected on Days 1, 2, 3, or 5 followed by multiplex cytokine analysis for IFNg, IL-2, IL-6, IL-1beta, IL-10 or TNFalpha. The data was quantified using MSD (Meso Scale Discovery) platform, following the manufacturer's protocol.

As shown in FIG. 23A, when plate-bound anti-TCRβV x BCMA bispecific molecule (Molecule H) or anti-CD3e antibodies (OKT3) were used to activate human PBMCs, the T cell cytokine IFNg was induced. With respect to IL-2 production, PBMCs activated with anti-TCRβV x BCMA bispecific molecule (Molecule H) resulted in increased IL-2 production (FIG. 23B) as compared to PBMCs activated with anti-CD3e antibodies (OKT3).

The production of cytokines IL-6, IL-1β, IL-10 and TNF-alpha which are associated with “cytokine storms” (and accordingly CRS) was also assessed under similar conditions. FIGS. 23C-E show that that while PBMCs activated with anti-CD3e antibodies demonstrate production of IL-1β (FIG. 23C), IL-6 (FIG. 23D), TNF-alpha (FIG. 23D) and IL-10 (FIG. 23E), no or little induction of these cytokines was observed with PBMCs activated with anti-TCRβV x BCMA bispecific molecule (Molecule H).

The data provided in this Example show that antibodies directed against TCR V(3 can, e.g., preferentially activate a subset of T cells, and do not result in induction of cytokines associated with cytokine storm or CRS.

Example 10: Cytokine and Chemokine Profile of Anti-TCRVb Antibodies

This Examples describes cytokines and chemokines secreted by PBMCs following activation by anti-TCR V(3 antibodies.

Human PBMCs were isolated from whole blood followed by solid-phase (plate-coated) stimulation with an anti-TCRβV antibodies (A-H.1, B-H.1), or a bispecific molecule comprising an anti-TCRVb antibody (Molecule H), an isotype control (BGM0122) or an anti-CD3e antibody (SP34), each at 100 nM. Supernatant was collected on Days 1, 2, 3, 4, 5, 6, 7 and 8 followed by multiplex analysis for the indicated cytokines or chemokines. The data was quantified using MSD (Meso Scale Discovery) platform, following the manufacturer's protocol. BGM0122 comprises the amino acid sequence of

(SEQ ID NO: 3283) METDTLLLWVLLLWVPGSTGDKTHTCPPCPAPELLGGPSVFLFPPKPKDT LMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTY RVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYT LPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDS DGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKGGG GSGGGGSGLNDIFEAQKIEWHE.

FIGS. 25A-25J, FIGS. 26A-2611, and FIGS. 27A-27L show the levels of cytokine and chemokine from PBMCs activated with the indicated antibodies.

As shown in FIG. 25A, when plate-bound anti-TCR VP antibodies or anti-CD3e antibodies (OKT3) were used to activate human PBMCs, the T cell cytokine IFNg was induced. With respect to IL-2 production, PBMCs activated with anti-TCR VP antibodies resulted in increased IL-2 production with delayed kinetics (FIG. 25B) as compared to PBMCs activated with anti-CD3e antibody (OKT3).

While IL-1beta (FIG. 25C), IL-6 (FIG. 25D), IL-10 (FIG. 25E), IL-4 (FIG. 25F), TNFalpha (FIG. 25G), IP-10 (FIG. 26C), IL-12-23p40 (FIG. 27D), IL-17A (FIG. 27G), and IL-1a (FIG. 2711), were induced by anti-CD3e antibody (OKT3), no or little induction of these cytokines or chemokines was observed with PBMCs activated with anti-TCRVb antibodies.

PBMCs activated with anti-TCR VP antibodies demonstrated induction of IL-13 (FIG. 251), IL-8 (FIG. 25J), Eotaxin (FIG. 26A), Eotaxin 3 (FIG. 26B), IL-18 (HA) (FIG. 26C), MCP-1 (FIG. 26E), MCP-4 (FIG. 26F), MDC (FIG. 26G), MIP1a (FIG. 2611), MIP1B (FIG. 27A), TARC (FIG. 27B), GM-CSF (FIG. 27C), IL-15 (FIG. 27E), IL-16 (FIG. 27F), and IL-15 (FIG. 271), IL-7 (FIG. 27J).

Example 11: Nanostring-Based Gene Expression Profiling of TCR Vb-Activated T Cells

This Example describes gene expression profiling of TCR Vβ-activated T cells to, e.g., uncover potential mechanisms or pathways underlying TCR Vβ activation of T cells.

In a first study, the anti-TCR V013.1 antibody A-H.1 was compared with an anti-CD3 antibody OKT3. Briefly, human PBMCs were isolated from whole blood. From isolated PBMCs, human CD3+ T cells were isolated using magnetic-bead separation (negative selection) (Miltenyi biotec) and activated by immobilized (plate-coated) anti-TCR V013.1 antibody (A-H.1) or anti-CD3 antibody (OKT3) at 100 nM for 6 days. Activated T-cells (from plate-coated) were then prepared for gene expression profiling (PanCancer IO 360™ Panel, nanoString), following manufacturer's protocol. Differential gene expression analysis was grouped by anti-TCR Vβ13.1 (A-H.1) vs anti-CD3 (OKT3) activated T-cells using the nSolver Analysis Software (Nanostring). Data shown in Table 15A are mean values from 3 donors. The differentially regulated genes shown in Table 15A have a p-value of 0.05 or less. In the fourth column of Table 15A showing fold changes in gene expression, a positive value indicates genes that are upregulated at the transcriptional level in TCR Vβ-activated T cells compared to OKT3-activated T cells, whereas a negative value indicates genes downregulated at the transcriptional level in TCR Vβ-activated T cells compared to OKT3-activated T cells.

TABLE 15A Summary of genes whose expression are preferentially regulated in TCR Vβ-activatedT cells compared to OKT3-activated T cells. TCR Vβ13.1 vs OKT3 Fold Probe Name Accession # NS Probe ID Change P value CCR2 NM_001123041.2 NM_001123041.2:743 −3.06 0.00019145 LIF NM_002309.3 NM_002309.3:1240 21.6 0.0003319 TCF7 NM_003202.2 NM_003202.2:2420 −8.38 0.00037035 PLA2G6 NM_001004426.1 NM_001004426.1:1954 −2.19 0.00043564 CD84 NM_001184879.1 NM_001184879.1:28 −3.81 0.00062413 ITGB2 NM_000211.2 NM_000211.2:520 −2.11 0.0012003 GZMK NM_002104.2 NM_002104.2:700 −11.09 0.00135083 HLA-DRB4 NM_021983.4 NM_021983.4:194 −5.75 0.00137591 CCR7 NM_001838.2 NM_001838.2:1610 −2.43 0.00165716 PDCD1 NM_005018.1 NM_005018.1:175 7.24 0.00195468 CD96 NM_005816.4 NM_005816.4:1355 −6.44 0.00221401 SELL NR_029467.1 NR_029467.1:1585 −5 0.00227156 NFATC4 NM_001136022.2 NM_001136022.2:2296 −2.75 0.0025171 CD8B NM_004931.3 NM_004931.3:440 −3.56 0.00302475 NLRC5 NM_032206.4 NM_032206.4:860 −2.27 0.00309164 CD1C NM_001765.2 NM_001765.2:750 8.62 0.0035729 HLA-B NM_005514.6 NM_005514.6:937 −1.81 0.00363669 NUP107 NM_020401.2 NM_020401.2:1002 1.64 0.00366886 CD3D NM_000732.4 NM_000732.4:110 −2.05 0.00401569 HDAC3 NM_003883.2 NM_003883.2:1455 −1.41 0.0042794 PRKCE NM_005400.2 NM_005400.2:1695 −1.86 0.00429076 HLA-DQB1 NM_002123.3 NM_002123.3:384 −5.71 0.00430297 AKT3 NM_181690.1 NM_181690.1:755 −2.98 0.00430433 VCAM1 NM_001078.3 NM_001078.3:2535 −23.93 0.00464703 CD53 NM_001040033.1 NM_001040033.1:835 −1.7 0.00507702 LRP1 NM_002332.2 NM_002332.2:4240 −2.22 0.00508974 CD28 NM_001243078.1 NM_001243078.1:2065 −1.73 0.00545641 OSM NM_020530.4 NM_020530.4:580 8.97 0.00558554 CLEC4A NM_194448.2 NM_194448.2:388 −1.7 0.0056661 MFGE8 NM_001114614.1 NM_001114614.1:328 −2.75 0.00633707 IFNAR2 NM_000874.3 NM_000874.3:631 −3.69 0.00659279 LTA NM_000595.2 NM_000595.2:885 6.53 0.00727884 ITGAE NM_002208.4 NM_002208.4:3405 −3.42 0.00779862 CXCR5 NM_001716.3 NM_001716.3:2618 4.38 0.00781195 CD6 NM_006725.3 NM_006725.3:1280 −1.38 0.00848703 ICOS NM_012092.2 NM_012092.2:640 1.74 0.00914866 NOS2A NM_153292.1 NM_153292.1:546 −2.29 0.0095337 CD1A NM_001763.2 NM_001763.2:1815 5.12 0.00956367 CD27 NM_001242.4 NM_001242.4:330 −3.41 0.00984676 KLRD1 NM_002262.3 NM_002262.3:542 −6.43 0.00998325 TARP NM_001003799.1 NM_001003799.1:560 −3.71 0.00998698 HLA-DPB1 NM_002121.4 NM_002121.4:931 −8.85 0.01064161 PTPRC NM_080921.3 NM_080921.3:258 −2.86 0.01124117 CD44 NM_001001392.1 NM_001001392.1:429 −2.07 0.01138242 SLAMF6 NM_001184714.1 NM_001184714.1:1032 −1.81 0.00175123 HLA-DMB NM_002118.3 NM_002118.3:20 −6.39 0.01184625 CD276 NM_001024736.1 NM_001024736.1:2120 6.22 0.01207813 MAGEA1 NM_004988.4 NM_004988.4:476 −2.93 0.01210408 HLA-DMA NM_006120.3 NM_006120.3:380 −5.75 0.1210789 EP300 NM_001429.2 NM_001429.2:715 −1.24 0.01228626 ADA NM_000022.2 NM_000022.2:1300 −2.97 0.01228787 ICAM1 NM_000201.2 NM_000201.2:2253 2.52 0.01290081 SIGIRR NM_021805.2 NM_021805.2:469 −4.46 0.01309473 TNF NM_000594.2 NM_000594.2:1010 4.6 0.01318389 IL1RAP NM_002182.2 NM_002182.2:460 2.77 0.01329693 CSF1 NM_000757.4 NM_000757.4:823 2.55 0.01373637 CD40LG NM_000074.2 NM_000074.2:1225 11.92 0.01376174 CYFIP2 NM_001037332.2 NM_001037332.2:4043 −1.38 0.01389707 MUC1 NM_001018017.1 NM_001018017.1:725 3.12 0.01399543 HLA-DRB3 NM_022555.3 NM_022555.3:698 −7.11 0.01404049 CD2 NM_001767.3 NM_001767.3:687 −1.53 0.01432842 IL2RG NM_000206.1 NM_000206.1:595 −1.82 0.01477006 HLA-A NM_002116.5 NM_002116.5:1000 −1.96 0.01454336 TXK NM_003328.1 NM_003328.1:800 −2.7 0.01590341 ITGA4 NM_000885.4 NM_000885.4:975 −3.59 0.01601785 DHX16 NM_001164239.1 NM_001164239.1:2490 1.41 0.0167432 CD3E NM_000733.2 NM_000733.2:75 −1.52 0.01736902 MR1 NM_001531.2 NM_001531.2:7695 −2.26 0.01744764 SMAD3 NM_005902.3 NM_005902.3:4220 −2.82 0.01751245 CCRL2 NM_003965.4 NM_003965.4:1110 −1.87 0.01834479 HRAS NM_005343.2 NM_005343.2:396 1.97 0.0187379 IL18R1 NM_003855.2 NM_003855.2:2025 2.36 0.01896204 CMA1 NM_001836.2 NM_001836.2:561 −1.96 0.01964938 PSMB7 NM_002799.2 NM_002799.2:420 1.53 0.01980367 BCL10 NM_003921.2 NM_003921.2:1250 −1.38 0.01981376 HLA-DRA NM_019111.3 NM_019111.3:335 −7.46 0.02026993 CD80 NM_005191.3 NM_005191.3:1288 4.18 0.02055337 PIK3CD NM_005026.3 NM_005026.3:2978 −1.23 0.02056576 ETS1 NM_005238.3 NM_005238.3:4625 −1.51 0.02083359 CHUK NM_001278.3 NM_001278.3:860 1.67 0.0217326 CCL5 NM_002985.2 NM_002985.2:280 −2.47 0.02195802 ITGAL NM_002209.2 NM_002209.2:3905 −3 0.02244779 TNFRSF18 NM_004195.2 NM_004195.2:445 −3.76 0.02330885 EIF2B4 NM_172195.3 NM_172195.3:1390 1.28 0.02349098 CD79A NM_001783.3 NM_001783.3:695 −4.47 0.02361746 ABCF1 NM_001090.2 NM_001090.2:850 1.31 0.02452054 CD37 NM_001774.2 NM_001774.2:535 −2.06 0.02476513 STAT5B NM_012448.3 NM_012448.3:200 −1.56 0.02495121 CSF2 NM_000758.2 NM_000758.2:475 11.38 0.0256982 STAT3 NM_139276.2 NM_139276.2:4535 −1.47 0.02629936 GZMA NM_006144.2 NM_006144.2:155 −2.46 0.02646368 C1R NM_001733.4 NM_001733.4:760 −3.1 0.02653879 MIF NM_002415.1 NM_002415.1:319 −1.38 0.02690018 CD46 NM_172350.1 NM_172350.1:365 −1.36 0.02725208 PIK3CG NM_002649.2 NM_002649.2:2125 −2.34 0.02762105 CFB NM_001710.5 NM_001710.5:2029 −2.59 0.02802998 IL3 NM_000588.3 NM_000588.3:130 13.37 0.02820076 TNFRSF13C NM_052945.3 NM_052945.3:789 −2.2 0.02835259 MRPS5 NM_031902.3 NM_031902.3:390 1.2 0.02849936 TUBB NM_178014.2 NM_178014.2:320 1.06 0.02874459 PECAM1 NM_000442.3 NM_000442.3:1365 −4.35 0.02901845 PVR NM_006505.3 NM_006505.3:604 2.28 0.0299334 AMICA1 NM_153206.2 NM_153206.2:620 −2.38 0.03034954 CD74 NM_001025159.1 NM_001025159.1:964 −3.28 0.0305419 ENTPD1 NM_001098175.1 NM_001098175.1:8830 −8.02 0.03085618 CD97 NM_078481.2 NM_078481.2:1370 −1.56 0.03086014 KLRK1 NM_007360.3 NM_007360.3:522 −4.16 0.03108504 HLA-DQA1 NM_002122.3 NM_002122.3:261 −5.51 0.03126291 CD247 NM_198053.1 NM_198053.1:1490 −1.88 0.03182703 IFNG NM_000619.2 NM_000619.2:970 5.98 0.03202586 SAA1 NM_199161.1 NM_199161.1:135 −2.35 0.03341258 TBX21 NM_013351.1 NM_013351.1:890 1.92 0.03359165 RORA NM_134261.2 NM_134261.2:1715 −2.57 0.03591525 MASP2 NM_139208.1 NM_139208.1:330 −1.65 0.03611762 CLU NM_001831.2 NM_001831.2:2340 −1.55 0.0369776 KLRB1 NM_002258.2 NM_002258.2:85 −7.43 0.03705134 RELA NM_021975.2 NM_021975.2:360 −1.26 0.03765981 SLAMF1 NM_003037.2 NM_003037.2:580 1.82 0.03768168 CD8A NM_001768.5 NM_001768.5:1320 −4.49 0.0380276 IL11RA NM_147162.1 NM_147162.1:400 −3.54 0.03855863 CD3G NM_000073.2 NM_000073.2:404 −1.44 0.03877635 JAK1 NM_002227.1 NM_002227.1:285 −1.84 0.4001383 SPN NM_003123.3 NM_003123.3:2345 −1.72 0.04035383 CXCR4 NM_003467.2 NM_003467.2:1335 −3.03 0.04122601 FAS NM_000043.3 NM_000043.3:90 −2.37 0.04150638 IL2 NM_000586.2 NM_000586.2:300 10.9 0.04175377 ITGA1 NM_181501.1 NM_181501.1:1875 −2.75 0.04213304 IGF1R NM_000875.2 NM_000875.2:455 −1.94 0.0424234 CLEC6A NM_001007033.1 NM_001007033.1:342 −2.83 0.04299769 RPS6 NM_001010.2 NM_001010.2:171 −1.36 0.04334091 MAPK11 NM_002751.5 NM_002751.5:1310 −1.98 0.04344288 REL NM_002908.2 NM_002908.2:225 −2.37 0.04382344 EOMES NM_005442.2 NM_005442.2:1670 −6.49 0.04442535 KLRG1 NM_005810.3 NM_005810.3:65 −3.52 0.04487411 IL2RA NM_000417.1 NM_000417.1:1000 3.4 0.0457568 IFNA17 NM_021268.2 NM_021268.2:291 −3.13 0.04595868 SH2D1B NM_053282.4 NM_053282.4:545 −1.44 0.04640447 CCL2 NM_002982.3 NM_002982.3:123 4.01 0.04660539 TXNIP NM_006472.1 NM_006472.1:255 −4.07 0.04695375 CXCL13 NM_006419.2 NM_006419.2:210 −65.05 0.04708191 CASP8 NM_001228.4 NM_001228.4:301 −1.42 0.04720592 MTMR14 NM_022485.3 NM_022485.3:720 −1.25 0.04798024 MAP3K5 NM_005923.3 NM_005923.3:1760 −1.62 0.04838454 ADORA2A NM_000675.3 NM_000675.3:1095 1.3 0.04872028 CCR5 NM_000579.1 NM_000579.1:2730 −4.01 0.04885927

In a second study, the multispecific anti-TCR Vβ13.1/anti-BCMA antibody Molecule H was compared with the anti-CD3 antibody OKT3. Purified T cells were stimulated with solid-phase anti-TCR Vβ antibody over 6 days with the anti-TCR Vβ antibody Molecule H or anti-CD3e antibody (OKT3) at 100 nM. Expanded T cells were collected by centrifugation followed by RNA extraction. Seven hundred and seventy eight (778) immunology-related genes were counted using the nCounter Technology (Nanostring) followed by gene expression analysis using nSolver analysis tools. The data described in this Example is representative of 3 donors.

Based on this analysis, a panel of genes were identified as being differentially regulated in TCR Vβ-activated T cells compared to OKT3-activated T cells (Table 15B). The differentially regulated genes shown in Table 15B have a p-value of 0.05 or less. For example, LIF, CD40LG, PDCD1, CXCR5, LTA, and CD80 are all upregulated at the transcriptional level in TCR Vβ-activated T cells compared to OKT3-activated T cells. GZMK, ENTPD1 (CD39), TCF7, CD96, HLA-DRB4, SIGIRR and SELL are downregulated at the transcriptional level in TCR Vβ-activated T cells compared to OKT3-activated T cells. TCR Vβ-activated T cells also expressed high levels of cytolytic effectors (e.g., IFNg, Granzyme B and perforin).

TABLE 15B Summary of genes whose expression are preferentially regulated in TCR Vβ-activated T cells compared to OKT3-activated T cells. Log2 Fold Gene Description Change P-Value LIF LIF Interleukin 6 Family Cytokine 4.65 0.0119 GZMK Granzyme K −3.65 0.0468 CD40LG CD40 Ligand 3.56 0.0082 ENTPD1 (CD39) Ectonucleoside Triphosphate Diphosphohydrolase 1 −3.53 0.0541 PDCD1 Programmed Cell Death 1 3.19 0.0257 TCF7 Transcription Factor 7 −3.1 0.00634 CXCR5 Chemokine receptor for CXCL13 3.05 0.0337 CD96 Transmembrane glycoprotein Ig superfamily receptor, −2.75 0.007 interacts with nectin and nectin-like proteins, including CD155/polio virus receptor (PVR) LTA Lymphotoxin Alpha 2.67 0.0082 HLA-DRB4 Major Histocompatibility Complex, Class 11, DR Beta 4 −2.66 0.0377 CD80 T cell costimulatory molecule 2.58 0.0425 SIGIRR Single Ig And TIR Domain Containing −2.37 0.0227 SELL Selection L −2.3 0.00634

Example 12: Binding Affinity of Affinity Matured Humanized Antibody A-H Antibodies

This Example describes the evaluation of binding affinity of affinity matured humanized Antibody A-H antibodies to recombinant protein TCRVB 6-5.

Antibody A-H humanized antibodies were affinity matured. The resulting affinity matured antibodies were tested for their binding affinity to TCRVB 6-5 as described below.

TCRVB 6-5 at 5 ug/mL was immobilized on a Biotin CAP Series S Sensor Chip to 60 RU. BJM0277 was diluted to 200 nM and then serially diluted two fold. Association was 120 seconds, and dissociation was 300 seconds. This assay was run in 1×HBS-EP+ Buffer pH 7.4 and 25C. The data was fit using a 1:1 binding model.

TCRVB 6-5 at 5 ug/mL was immobilized on a Biotin CAP Series S Sensor Chip to 60 RU. A-H.45 was diluted to 50 nM and then serially diluted two fold. Association was 120 seconds, and dissociation was 300 seconds. This assay was run in 1×HBS-EP+ Buffer pH 7.4 and 25C. The data was fit using a 1:1 binding model. A-H.45 is an improved yeast clone (TCRvB/CD19 bispecific) and contains a mutation (G to V) at the last residue in framework 3, just before HCDR3. The affinity is 35-fold greater than the BJM0277 (Table 16).

TCRVB 6-5 at 5 ug/mL was immobilized on a Biotin CAP Series S Sensor Chip to 60 RU. A-H.52 was diluted to 50 nM and then serially diluted two fold. Association was 120 seconds, dissociation was 300 seconds. This assay was run in 1×HBS-EP+ Buffer pH 7.4 and 25C. The data was fit using a 1:1 binding model. A-H.52 is a phage clones and is a monovalent scFv. A-H.52 has two mutations on CDRH1. The affinity of A-H.52 is 20-fold greater than BJM0277 (Table 16).

TCRVB 6-5 at 5 ug/mL was immobilized on a Biotin CAP Series S Sensor Chip to 60 RU. A-H.53 was diluted to 50 nM and then serially diluted two fold. Association was 120 seconds, dissociation was 300 seconds. This assay was run in 1×HBS-EP+ Buffer pH 7.4 and 25C. The data was fit using a 1:1 binding model. A-H.53 (phage clone) affinity is in the pM range (Table 16). The affinity of A-H.53 is 200-fold greater than BJM0277 (Table 16).

TCRVB 6-5 at 5 ug/mL was immobilized on a Biotin CAP Series S Sensor Chip to 60 RU. A-H.54 was diluted to 50 nM and then serially diluted two fold. Association was 120 seconds, dissociation was 300 seconds. This assay was run in 1×HBS-EP+ Buffer pH 7.4 and 25C. The data was fit using a 1:1 binding model. A-H.54 (phage clone) affinity is 17-fold greater than BJM0277 (Table 16).

TABLE 16 Summary of affinity maturation of anti-TCRVb antibodies Target: Construct TCRVβ 6-5 BJM0277   35 nM A-H.45 1.08 Nm A-H.52 1.76 nM A-H.53  165 pM A-H.54 2.22 nM

Example 13: Therapeutic Efficacy of CD19/TCRvB Bispecific Molecules in Subcutaneous Human Tumor Xenograft Models

This Example demonstrates the in vivo efficacy of a CD19/TCRvB Bispecific molecule in a subcutaneous human tumor animal model.

On day 1 of the study 1×106 cells of the human cancer cell line Raji, stably expressing firefly luciferase (Raji-luc) were subcutaneously injected in the right dorsal flank of female NOD/SCID/IL-2Rγnull (NSG) mice. On day 3, 10×106 human PBMCs were transplanted into mice by injection into the peritoneal cavity.

Antibody treatment started at day 10, when tumors had reached a mean tumor volume (TV) of 80 mm3. Mean TV of each group was not statistically different from any other group at start of treatment. Mice were treated with 0.2 mg/kg, 1 mg/kg and 5 mg/kg of CD19/TCRvB bispecific molecule every three days for a total of 7 doses by intravenous bolus injection.

Tumor volume (TV) was measured every 3 days by calipers and progress evaluated by intergroup comparison of TV. Tumor growth inhibition T/C [%] was calculated as T/C[%]=100×(mean TV of analyzed group)/(mean TV of vehicle group).

Results are shown in Table 17 and FIG. 28. Treatment with the CD19/TCRvB Bispecific molecule inhibited tumor growth compared to vehicle control treatment (FIG. 28). The results demonstrate that the CD19/TCRvB bispecific molecule inhibits tumor growth and has anti-tumor activity.

TABLE 17 Mean tumor volume and tumor growth inhibition (T/C) at days 10 to 28. Dose group Data D10 D13 D16 D19 D23 D25 D28 Vehicle TV (mm3) 84 241 566 802 1577 2161 2478 T/C[%] 100 100 100 100 100 100 100 0.2 mg/kg CD19/TCRvB TV (mm3) 82 169 460 643 967 946 875 T/C[%] 98 70 81 80 61 44 35 1 mg/kg CD19/TCRvB TV (mm3) 82 122 147 307 482 469 406 T/C[%] 98 51 26 38 31 22 16 5 mg/kg CD19/TCRvB TV (mm3) 79 160 200 381 510 409 382 94 66 35 48 32 19 15

Example 14: Therapeutic Efficacy of CD19/TCRvB Bispecific Molecules in Human Tumor Xenograft Models

This Example demonstrates the in vivo efficacy of a CD19/TCRvB Bispecific molecules in a xenograft animal model.

On day 1 of the study 10×106 human PBMCs were transplanted into NOD/SCID/IL-2Rγnull (NSG) mice by injection into the peritoneal cavity.

On day 7, 1×106 cells of the human cancer cell line Raji, stably expressing firefly luciferase (Raji-luc) were intravenously injected into NOD/SCID/IL-2Rγnull (NSG) mice. Control animals were injected with 10×106 cells of the CD19 negative human cancer cell line K562 stably expressing firefly luciferase (K562-luc). These animals were used to assess specific killing ability of CD19/TCRvB molecules. Antibody treatment started at day 16, when tumor engraftment had reached a mean bioluminescence flux level of 4×107 photons/s. Mean Flux level of each group was not statistically different from any other group at start of treatment. Mice were treated with 1 mg/kg and 5 mg/kg of CD19/TCRvB bispecific molecule every three days for a total of 6 doses by intravenous bolus injection.

Tumor burden was measured weekly by bioluminescence imaging and progress evaluated by intergroup comparison of total bioluminescence flux (Total Flux). Tumor growth inhibition T/C [%] was calculated as T/C[%]=100×(mean Total Flux of analyzed group)/(mean Total Flux of vehicle group).

The results for Raji-luc engrafted animals are shown in Table 18 and FIG. 29A and results for K562-luc engrafted animals are shown in Table 19 and FIG. 29B. The results demonstrate that the CD19/TCRvB bispecific molecule inhibits tumor growth and has anti-tumor activity (FIG. 29A and Table 18).

TABLE 18 Mean tumor burden (Total Flux) and tumor growth inhibition (T/C) at days 16 to 37 in animals engrafted with Raji-luc cells Dose group Data D16 D23 D30 D37 Vehicle Total Flux (p/s) 4.26E+07 5.92E+07 5.77E+08 4.23E+09 T/C [%] 100 100 100 100 1 mg/kg CD19/TCRvB Total Flux (p/s) 4.05E+07 2.66E+07 5.03E+07 5.42E+08 T/C [%] 95.0 44.9 8.7 12.8 5 mg/kg CD19/TCRvB Total Flux (p/s) 4.18E+07 3.10E+07 2.37E+07 1.44E+08 T/C [%] 98.0 52.3 4.1 3.4

TABLE 19 Mean tumor burden (Total Flux) and tumor growth inhibition (T/C) at days 16 to 30 in animals engrafted with K562-luc cells Dose group Data D16 D23 D30 Vehicle Total Flux (p/s) 2.98E+07 9.94E+08 2.40E+10 T/C [%] 100 100 100 5 mg/kg Total Flux (p/s) 2.00E+07 1.22E+09 3.82E+10 CD19/TCRvB T/C [%] 67.0 122.4 159.4

Example 15: Therapeutic Efficacy of BCMA/TCRvB Bispecific Molecules in Human Tumor Xenograft Models

This Example demonstrates the in vivo efficacy of a BCMA/TCRvB Bispecific molecule in a xenograft animal model.

On day 1, 20×106 cells of the human cancer cell line RPMI-8226, stably expressing firefly luciferase (RPMI-8226-luc) were intravenously injected into NOD/SCID/IL-2Rγnull (NSG) mice. On day 11, 10×106 human PBMCs were transplanted into mice by injection into the peritoneal cavity. Antibody treatment started at day 17, when tumor engraftment had reached a mean bioluminescence flux level of 4×107 photons/s. Mice were treated with 0.5 mg/kg of a molecule bivalent for both BCMA and TCRvB (2×2 molecule) and 0.5 mg/kg of a molecule bivalent for BCMA and monovalent for TCRvB (2×1 molecule) once a week for a total of 2 doses by intravenous bolus injection.

Tumor burden was measured weekly by bioluminescence imaging and progress evaluated by intergroup comparison of total bioluminescence flux (Total Flux). Tumor growth inhibition T/C [%] was calculated as T/C[%]=100×(mean Total Flux of analyzed group)/(mean Total Flux of vehicle group).

Results of these studies are shown in Table 20 and FIG. 30. Treatment with the BCMA/TCRvB Bispecific molecule inhibited tumor growth compared to vehicle control treatment (FIG. 29). The results demonstrate that the BCMA/TCRvB bispecific molecule inhibits tumor growth and has anti-tumor activity.

TABLE 20 Mean tumor burden (Total Flux) and tumor growth inhibition (T/C) at days 16 to 30. Dose group Data D16 D23 D30 Vehicle Total Flux (p/s) 3.71E+06 6.04E+06 7.29E+06 T/C [%] 100 100 100 0.5 mg/kg Total Flux (p/s) 7.33E+06 6.30E+06 1.13E+06 BCMA/TCRvB 2 × 2 T/C [%] 197.7 104.3 15.5 0.5 mg/kg Total Flux (p/s) 3.66E+06 3.15E+06 5.65E+05 BCMA/TCRvB 2 × 1 T/C [%] 98.8 52.1 7.8

Example 16: Expression and Purification of Antibody Constructs

Construction of the Plasmids

The DNA encoding the protein sequences was optimized for expression in Cricetulus griseus, synthesized, and cloned into the pcDNA3.4-TOPO (Life Technologies A14697) using Gateway cloning. All constructs contained an Ig Kappa leader sequence METDTLLLWVLLLWVPGSTG (SEQ ID NO: 3288).

Expression and Purification

The plasmids were co-transfected into either Expi293 cells (Life Technologies A14527) or ExpiCHO cells (Life Technologies A29127). Transfections were performed using 1 mg of total DNA for a multispecific construct with a 1:1 heavy chain ratio and 3:2 light chain to heavy chain ratio if applicable. Transfection in Expi293 cells was done using linear 25,000 Da polyethylenimine (PEI, Polysciences Inc 23966) in a 3:1 ratio with the total DNA. The DNA and PEI were each added to 50 mL of OptiMem (Life Technologies 31985088) medium and sterile filtered. The DNA and PEI were combined for 10 minutes and added to the Expi293 cells with a cell density of 1.8-2.8×106 cells/mL and a viability of at least 95%. The ExpiCHO transfection was performed according to the manufacturer's instructions. Expi293 cells were grown in a humidified incubator at 37° C. with 8% CO2 for 5-7 days after transfection and ExpiCHO cells were grown for 14 days at 32° C. with 5% CO2. The cells were pelleted by centrifugation at 4500×g and the supernatant was filtered through a 0.2 μm membrane. Protein A resin (GE 17-1279-03) was added to the filtered supernatant and incubated for 1-3 hours at room temperature. The resin was packed into a column, washed with 3×10 column volumes of Dulbecco's phosphate-buffered saline (DPBS, Life Technologies 14190-144). The bound protein was eluted from the column with 20 mM citrate, 100 mM NaCl, pH 2.9. When necessary, the proteins were further purified using ligand affinity and/or size exclusion chromatography on a Superdex 200 column with a running buffer of DPBS.

Example 17: Humanization of Anti-TRBV5-5 Antibody Clone Antibody C

The germline for the mouse anti-TCRvbeta antibody clone Antibody C VH and VL were assigned using IMGT nomenclature, with CDR regions defined by a combined Kabat and Chothia classification. SEQ ID NO: 232 and SEQ ID NO: 233 are the Antibody C VH and VL sequences respectively where the VH germline is mouse IGHV2-6-7*01 and the VL germline is mouse IGKV10-94*02. The method applied to humanize Antibody A described in Example 1 was used to humanize Antibody C. The Antibody C VH was humanized into human IGHV2-26*01, IGHV2-70*04, IGHV4-4*02, IGHV2-5*09, IGHV2-5*08, IGHV4-34*09, IGHV4-59*01, IGHV4-59*07, IGHV4-61*02, IGHV4-38-2*01, IGHV4-31*01, IGHV3-49*04, IGHV3-49*02, IGHV4-4*07, IGHV3-49*05, IGHV4-34*10, IGHV4-28*04, IGHV3-72*01, IGHV3-15*07, IGHV6-1*01, IGHV3-7*01, IGHV4-34*01, IGHV3-33*02, IGHV3-48*02, IGHV3-23*03, IGHV3-21*01, IGHV3-73*01, IGHV3-30*02, IGHV3-7*01, IGHV3-43*01, and IGHV3-53*03 and the Antibody C VL was humanized into human IGKV1D-43*01, IGKV1-27*01, IGKV1-17*02, IGKV1-17*01, IGKV1-5*01, IGKV4-1*01, IGKV3-7*02, IGKV3-7*01, IGKV2-29*02, IGKV6D-41*01, IGKV2-28*01, IGKV2-40*01, IGKV3-15*01, IGKV2-24*01, IGKV6-21*01, IGKV2D-26*01, and IGKV2D-26*03.

SEQ ID NOs: 3040-3089 are the Antibody C humanized heavy chains and SEQ ID NOs: 3000-3039 are the Antibody C humanized light chains (as described in Table 10).

Example 18: Humanization of TRBV10-1, TRBV10-2, and TRBV10-3 Antibody Clone Antibody D

The germline for the mouse anti-TCRvbeta antibody clone Antibody D VH and VL were assigned using IMGT nomenclature, with CDR regions defined by a combined Kabat and Chothia classification. SEQ ID NO: 3183 and SEQ ID NO: 3184 are the Antibody D VH and VL sequences respectively where the VH germline is mouse IGHV5-6*01 and the VL germline is mouse IGKV4-59*01.

The method applied to humanize Antibody A described in Example 1 was used to humanize Antibody D. The Antibody D VH was humanized into human IGHV3-30*03, IGHV3-30*02, IGHV3-7*01, IGHV3-21*01, IGHV3-23*04, IGHV3-30*15, IGHV3-48*02, IGHV3-53*04, IGHV3-23*03, IGHV3-53*03, IGHV3-53*01, IGHV3-9*01, IGHV3-30*13, IGHV3-20*01, IGHV3-43D*03, IGHV3-43*02, IGHV3-43*01, IGHV3-53*02, IGHV3-13*01, IGHV3-38-3*01, IGHV3-9*03, IGHV3-64D*06, IGHV3-33*02, IGHV3-11*03, IGHV3-64*02, IGHV3-64*01, IGHV3-64*03, IGHV3-7*01, IGHV3-35*01, IGHV3-13*02, IGHV3-38*02, and IGHV3-38*01 and the Antibody D VL was humanized into human IGKV3-11*01, IGKV1-13*02, IGKV1-9*01, IGKV6-21*01, IGKV1D-43*01, IGKV3-11*01, IGKV3D-11*02, IGKV1-17*03, IGKV3D-20*01, IGKV3-20*01, IGKV1D-16*01, IGKV4-1*01, IGKV2-28*01, IGKV2-40*01, IGKV2-29*02, IGKV2-29*01, IGKV1D-42*01, IGKV2-24*01, and IGKV5-2*01. SEQ ID NOs: 3225-3274 are the Antibody D humanized heavy chains and SEQ ID NOs: 3185-3224 are the Antibody D humanized light chains (as described in Table 12).

Example 19: Humanization of TRBV5-5 and TRBV5-6 Antibody Clone Antibody E

The germline for the mouse anti-TCRβ antibody clone Antibody E VH and VL were assigned using IMGT nomenclature, with CDR regions defined by a combined Kabat and Chothia classification. SEQ ID NO: 3091 and SEQ ID NO: 3092 are the Antibody E VH and VL sequences respectively where the VH germline is mouse IGHV1-82*01 and the VL germline is mouse IGKV3-5*01.

The method applied to humanize Antibody A described in Example 1 was used to humanize Antibody E. The Antibody E VH was humanized into human IGHV1-69*08, IGHV1-3*02, IGHV1-18*03, IGHV1-3*01, IGHV1-18*01, IGHV1-2*06, IGHV1-2*01, IGHV1-2*06, IGHV1-8*01, IGHV7-4-1*02, IGHV1-58*02, IGHV5-51*01, IGHV7-4-1*04, IGHV7-81*01, IGHV5-51*04, IGHV5-51*01, IGHV1-45*03, IGHV3-49*04, IGHV3-49*02, IGHV3-49*05, IGHV4-4*02, IGHV3-49*05, IGHV3-73*01, IGHV4-4*02, IGHV3-15*07, IGHV3-15*02, IGHV3-72*01, IGHV4-59*07, IGHV4-31*01, IGHV4-31*02, IGHV3-30*15, IGHV3-21*01, IGHV3-7*01, IGHV4-28*01, IGHV4-28*02, IGHV3-30*08, IGHV3-30*05, and IGHV3-30*01 and the Antibody E VL was humanized into human IGKV4-1*01, IGKV3-11*01, IGKV3-20*02, IGKV3-11*01, IGKV1-13*02, IGKV3D-11*01, IGKV3D-20*02, IGKV1-13*02, IGKV3D-20*01, IGKV1-9*01, IGKV3D-15*03, IGKV3-15*01, IGKV1-5*01, IGKV2D-29*01, IGKV3-7*02, IGKV1-9*01, IGKV2-28*01, IGKV2-40*01, IGKV2D-29*02, IGKV3-7*01, IGKV2-30*01, IGKV2-24*01, IGKV6D-41*01, IGKV1D-42*01, IGKV2D-26*01, IGKV2D-26*03, and IGKV5-2*01. SEQ ID NOs: 3133-3182 are the Antibody E humanized heavy chains and SEQ ID NOs: 3093-3132 are the Antibody E humanized light chains (as described in Table 11).

Example 20: In Vitro Cytotoxicity of an Anti-TCRVb/CD19 Antibody Molecule and an Anti-TCRVb/BCMA Antibody Molecule

Anti-TCR/Anti-CD19 Dual Targeting Antibody Molecule

Human PBMCs were isolated from whole blood. From isolated PBMCs, human CD3+ T cells were isolated using magnetic-bead separation (negative selection) (Miltenyi biotec) and activated by immobilized (plate-coated) anti-TCR V013.1 (A-H.1) at 100 nM for 6 days. Activated T-cells (from plate-coated) were then transferred and expanded in tissue culture flask in the presence of human IL-2 at a concentration of 50 U/ml for two additional days. Expanded TCR V013.1+ cells were washed and co-cultured in the presence of CD19-expressing Raji cells (target cells) at an E:T ratio of 5:1 and serial diluted concentrations of T-cell engager bispecific antibodies including, anti-TCR V013.1/CD19 (Molecule F), anti-CD3/CD19, and anti-TCR Vβ13.1 (A-H.1, serving as control) for 24 hours. Post 24 hours, cell co-culture supernatants were collected and quantified for specific target cell death. Target cells (Raji cells) are a KILR-retroparticles reporter cell assay (DiscoverX). KILR-Raji target cells are engineered to stably express a protein tagged with enhanced ProLabel (ePL), a β-gal reporter fragment, using the KILR Retroparticles, and when the membrane of the target cells is compromised due to cell death, the target cells will release the tagged protein into the media. This KILR reporter protein can be detected in the media/supernatant by the addition of detection reagents containing the enzyme acceptor (EA) fragment of the β-gal reporter. This leads to the formation of the active β-gal enzyme which hydrolyzes the substrate to give a chemiluminescent output (RLU). Percentage (%) of target cell death is calculated using the following formula:


(RLUTreatment−RLUNo Treatment)/(RLUMaximum Lysis−RLUNo Treatment)×100

Data shown in FIG. 31A are mean values from 4 donors.

Anti-TCR/Anti-BCMA Dual Targeting Antibody Molecule

Human PBMCs were isolated from whole blood. From isolated PBMCs, human CD3+ T cells were isolated using magnetic-bead separation (negative selection) (Miltenyi biotec) and activated by immobilized (plate-coated) anti-TCR V013.1 (A-H.1) at 100 nM for 6 days. Activated T-cells (from plate-coated) were then transferred and expanded in tissue culture flask in the presence of human IL-2 at a concentration of 50 U/ml for two additional days. Expanded TCR V013.1+ cells were washed and co-cultured in the presence of BCMA-expressing RPMI8226 cells (target cells) at an E:T ratio of 5:1 and serial diluted concentrations of T-cell engager bispecific antibodies including, anti-TCR Vβ13.1/BCMA (Molecule G), anti-CD3/BCMA, and anti-TCR V013.1 (A-H.1, serving as control) for 24 hours. Post 24 hours, cell co-culture supernatants were collected and quantified for specific target cell death. Target cells (RPMI8226 cells) are a KILR-retroparticles reporter cell assay (DiscoverX). KILR-RPMI8226 target cells are engineered to stably express a protein tagged with enhanced ProLabel (ePL), a β-gal reporter fragment, using the KILR Retroparticles, and when the membrane of the target cells is compromised due to cell death, the target cells will release the tagged protein into the media. This KILR reporter protein was detected and percentage (%) of target cell death was calculated as described above. Data shown in FIG. 31B are mean values from 4 donors.

Example 21: Cytokine Profile of an Anti-TCRVb/BCMA Antibody Molecule

This Example describes cytokines secreted by PBMCs following activation by the anti-TCR V0/anti-BCMA antibody Molecule H. For comparison, activation by an anti-TCR beta constant 1 (TRBC1) antibody Antibody F was also analyzed.

Briefly, human PBMCs were isolated from whole blood followed by solid-phase (plate-coated) stimulation with Molecule H or Antibody F at 100 nM. Supernatant was collected on Days 1, 2, 3, and 5 (for Molecule H) or Days 2 and 5 (for Antibody F) followed by multiplex cytokine analysis for IFNγ, IL-2, IL-1β, IL-6, IL-10, and TNFα, quantified using MSD (Meso Scale Discovery) platform, following the manufacturer's protocol.

As shown in FIGS. 32A-32F and 33A-33F, the cytokine profile of the anti-TCR V0/anti-BCMA antibody Molecule H is different from that of the anti-CD3 antibody OKT3 or the anti-TRBC1 Antibody F.

Example 22: Kinetics of T Cell Expansion Following TCRβV 6-5 Stimulation

To assess the kinetics and absolute count of anti-TCRβv 6-5 expanded T cells—either PBMCs or purified T cells were stimulated with plate-immobilized anti-TCRvb 6-5 antibody over 8 days with a T cell-activating antibody at 100 nM. T cell activating antibodies tested included: i) anti-TCRvb 6-5 v1 antibody; ii) anti-TCRvb 6-5 v2; iii) OKT3 (anti-CD3ε antibody); iv) SP34-2 (anti-CD3ε antibody); and v) IgG1 N297A (isotype control). Cell pellets were collected each day and stained for CD3, CD4, CD8 and TCRvb 6-5 for flow analysis.

TCRvb 6-5+ T cell expansion over 8 days using anti-TCRvb 6-5 v1 is shown in FIG. 34, as assessed by flow cytometry. The data is for a single representative donor; and similar results were seen with PBMCs from two other independent donors. FIG. 36 further shows the specific expansion of TCRvb 6-5+ CD4+ T cells and TCRvb 6-5+ CD8+ T cells by TCRvb 6-5 v1. In contrast, there was no specific TCRvb 6-5+ T-cell expansion by OKT3 (FIG. 35; FIG. 37).

FIGS. 38A and 38B show selective expansion of TCRβV 6-5+ T cells in human PBMCs (FIG. 38A) and purified T cells (FIG. 38B).

FIGS. 39A-41 shows that anti-TCRβV and anti-CD3ε antibodies expand T cells in a PBMC culture (FIGS. 39A and 39B) or a purified T cell culture (FIGS. 40A and 40B)) to comparable levels after 8 days, as measured by both relative count of TCRVB 6-5+ T cells (FIGS. 39A-40B) and relative count of total CD3+ T cells (FIGS. 39A-41).

Example 23: Activated TCRvb 6-5+ T Cells Exert Cytolytic Function

To assess the ability of T cells activated/expanded with anti-TCRVβ to mediate tumor cell lysis—purified T cells were stimulated over 6 days with an immobilized T cell-activating antibody at 100 nM. T cell activating antibodies tested included: i) TCRvb 6-5 v1 antibody; ii) OKT3 (anti-CD3ε antibody); or iii) IgG1 N297A (isotype control). Target cells (RPMI-8226 cells) were added on each day and incubated with the activated T cells at an initial effector T cell:target (E:T) cell ratio of 5:1 for 48 hours. Quantification of target cell lysis was measured using CFSE/CD138 and DRAQ7 FACS staining. Three different T cell donors were used (donor 6769, donor 9880, donor 54111). The data shows that the kinetics of target cell lysis by TCRVb 6-5 v1 activated T cells correlates with the expansion of TCRvb 6-5+ T cells (FIG. 42).

To further assess target cell lysis OKT3 or TCRvb 6-5 v1 antibodies were immobilized (plate-coated) with a ½ log serial dilution from a top dose concentration of 100 nM for purified T-cell (pan CD3 isolated) activation. The purified T-cells were stimulated with the activation plate for 0 (i.e. without antibody preactivation) to 4 (i.e. with antibody preactivation) days prior to addition of the target cells. Target cells (RPMI8226) were added to the activation plate (at an initial E:T cell ratio, 5:1) for up to 6-days (i.e. for plate 0, E:T coculture for 6-days, and for plate 4, E:T coculture for 2-days) followed by target cell lysis quantification via CFSE/CD138 and DRAQ7 FACS staining. The data shows that without T-cell preactivation, approximately 3% of Vb cells were able to kill target cells at day 6 (at higher concentration) (FIG. 43A); and with T-cell preactivation, approximately 25% of Vb cells were able to kill target cells at day 6 (the killing curve is shifted to the left) (FIG. 43B). TCRvb 6-5 v1 activated T cells exhibit comparable maximal target cell lysis when compared to anti-CD3ε when T cells are preactivated for 4 days (FIG. 44). At 100 nM, TCRvb 6-5 v1 activation shows comparable killing of target cells to anti-CD3ε activation (FIG. 45) (preactivation between 4-6 days depending on the donor and the cultures cultured for 48 h in presence of target cells).

Example 24: Assessing TCRvb Downregulation/Internalization by Anti-TCRvb 6-5 Antibody

To assess the effect anti-TCRvb 6-5 mediated T cell activation has on cell surface expression of TCRvb—purified T cells were stimulated over 8 days with the indicated T cell-activating antibody at 100 nM (plate bound). T cell activating antibodies included: i) anti-TCRvb 6-5 v1 antibody; or ii) SP34-2 (anti-CD3ε antibody). Cell pellets were collected each day and stained for CD3, CD4, CD8 and TCRβV 6-5 for flow cytometry analysis. A total of three donors were tested, each showing similar results.

The results show that both anti-CD3ε and anti-TCRvb antibodies activated CD4+ T cells (FIG. 46) and activated CD8+ T cells (FIG. 47) display reduced CD3c cell surface expression; whereas, TCRvb 6-5 cell surface expression on CD4+ T cells (FIG. 48) and CD8+ T cells (FIG. 49) remains detectable post T cell activation. The results show that the CD3c subunit is downregulated/internalized in T cells activated by either anti-CD3ε or anti-TCRvb antibodies; while TCRvb 6-5 remains detectable post T cell activation. Additionally, CD4 and CD8 staining did not show any signs of downmodulation of these receptors by either antibody.

Example 25: Cynomolgus Cross Reactivity of Anti-TCRβV Antibodies

To assess the cross reactivity of anti-TCRβV antibodies for cynomolgus TCRβV clonotype—fresh and cryopreserved cynomolgus PBMCs were cultured in complete media (RPMI with 10% FBS) in tissue culture treated flat bottom 96 well plates precoated with anti-TCRβV 6-5 v1 or anti-CD3ζ antibodies at 100 nM concentration. Negative control or unstimulated wells received PBS alone. TCRβV 6-5 expression was evaluated after 6 days in culture using CytoFlex flow cytometer (Beckmann Coulter) and imaged. Two donors samples were used: Donor DW8N—fresh PBMC sample, male, age 8, weight 7.9 kgs (data presented in FIG. 50A); Donor G709—cryopreserved sample, male, age 6, weight 4.7 kgs (data presented in FIG. 50B). The data show that cynomolgus T cells were activated and expanded by the anti-TCRβV 6-5 v1 (FIG. 50A and FIG. 50B). Fresh cynomolgus PBMCs from donor DW8N that had shown TCRvb 6-5 expansion were cryopreserved and after a week in cryopreservation, the cells were thawed and stimulated using anti-CD3ξ and anti-TCRvb 6-5 v1 for seven days. Cluster formation and expansion were both reproducible as shown in FIG. 51.

Example 26: No Activation of γδ T Cells by Anti-TCRβV Antibodies

To determine if anti-TCRvb antibodies are able to activate γδ T cells—γδ T cells were purified from human PBMCs via magnetic bead separation. γδ T cells were immobilized on plate-coated anti-CD3ε (SP34-2) or anti-TCRvb 6-5 (anti-TCRvb 6-5 v1) antibodies for 24 hours and analyzed for CD69 and CD25 expression by flow cytometry. Supernatants were collected post activation 2, 5, and 7 days, and analyzed for cytokines using Meso Scale Discovery (MSD) assay. FACS gating/staining on PBMCs was conducted prior to γδ T cell purification showing that γδ T cells are vβ6-5 negative (Donor 12657—gating for γδ T and TCRβ 6-5 based on FMO) (FIG. 52). FACS gating/staining on purified γδ T cell was conducted showing that purified γδ T cells are vβ6-5 negative (Donor 12657—gating for γδ T and TCRβ 6-5 based on FMO) (FIG. 53). As shown in FIG. 54, the anti-TCR Vβ6-5 antibody (anti-TCRvb 6-5 v1) did not activate γδ T cells; while the anti-CD3ε antibody (SP34-2) did. The cytokine analysis showed that anti-TCRβV 6-5 v1 does not induce cytokine release by γδ T cells, cytokines analyzed include IFNγ, TNFα, IL-2, IL-17A, IL-1α, IL-1β, IL-6, and IL-10 (FIG. 55A-55H).

Example 27: Polyclonal T Cell Expansion by Anti-TCRVβ Antibodies

To assess the ability of anti-TCRVβ antibodies to induce polyclonal T cell expansion—human CD3+ T cells were isolated using magnetic-bead separation (negative selection) and activated with immobilized (plate-coated) anti-TCRβV 6-5 v1 at 100 nM for 6 days. The expanded T cell population was washed and lysed using Takara single cell lysis buffer for SMART(er) TCR cDNA synthesis and sequencing. TCR sequencing was carried out and absolute counts and relative representation of the different TCR alpha V and J segments and TCR beta V, D, and J segments were determined, as well as the different variants of each of them that arise from Artemis/TdT activity during the V(D)J recombination, and that correspond to unique clones of T cells. FIG. 56 shows the relative representations of all TCR alpha V segments (TRAV group of genes) and their variants (top), all TCR beta V segment 6-5 variants (TRBV6-5 gene) (bottom left), and all TCR beta V segments and variants excluding 6-5 (bottom right). The data show that the anti-TCRVβ antibody stimulation does not induce proliferation of specific T cell clones within the TRBV6-5 positive population, as the relative difference in clonal representation in that population is comparable to the TRBV6-5 negative population as well as total TRAV usage.

Example 28: T Cells Expanded by Anti-TCRβV Represent a Novel Subset of Recently Activated Effector T Cells

To assess the phenotype of anti-TCRβV expanded T cells—purified T cells were stimulated with solid-phase anti-TCRβV antibody over 8 days with the indicated T cell-activating antibody at 100 nM: i) anti-TCRvb 6-5 v1 antibody; ii) anti-TCRvb 6-5 v2; iii) OKT3 (anti-CD3ε antibody); or iv) IgG1 N297A (isotype control). T-cell subsets were identified by FACS staining for specific surface markers for: Naïve T cell (CD4/CD8+, CD45RA+, CCR7+); T stem cell memory (TSCM; CD4/CD8+, CD95+, CD45RA+, CCR7+); T central memory (TCM; CD4/CD8+, CD95+, CD45RA−, CCR7+); T effector memory (TEM; CD4/CD8+, CD95+, CD45RA−, CCR7−); T effector memory re-expressing CD45RA (TEMRA; CD4/CD8+, CD95+, CD45RA+, CCR7−); and CD27, CD28, 4-1BB, OX40, and ICOS. Data is representative of more than 5 independent experiments.

The data shows that CD4+ T cells expanded by anti-TCR Vβ antibody (FIG. 57A), but not OKT3 (FIG. 57B), share phenotypic markers with the TEMRA subset. Likewise, the data shows that CD4+ T cells expanded by anti-TCR Vβ antibody (FIG. 58A), but not OKT3 (FIG. 58B), share phenotypic markers with the TEMRA subset. Further analysis of PD1 expression showed anti-TCR Vβ activated CD4+ T cells (FIG. 59A) and CD8+ T cells (FIG. 59B) display increased PD1 expression relative to anti-CD3ε activated CD4+ T cells (FIG. 59A) and CD8+ T cells (FIG. 59B). These anti-TCR Vβ activated CD4+ T cells (FIG. 60A) (PD-1+ TEMRA phenotype) and anti-TCR Vβ activated CD8+ T cells (FIG. 60B) (PD-1+ TEMRA phenotype) show Ki-67 enriched phenotype relative to anti-CD3ε activated CD4+ T cells (FIG. 60A) and CD8+ T cells (FIG. 60B).

Further analysis of CD57 expression showed anti-TCR Vβ activated CD8+ T cells (FIG. 61A) do not display increased CD57 expression relative to anti-CD3ε activated CD8+ T cells (FIG. 61B). Likewise, analysis of CD27 and CD28 expression showed anti-TCR Vβ activated CD4+ T cells (FIG. 62 top) and anti-TCR Vβ activated CD8+ T cells (FIG. 62 bottom) do not display increased CD27 and CD28 expression relative to anti-CD3ε activated CD8+ T cells (FIG. 62).

Further analysis of OX40, 41BB, and ICOS expression showed anti-TCR Vβ activated CD4+ T cells (FIG. 63 top) and anti-TCR Vβ activated CD8+ T cells (FIG. 63 bottom) display increased OX40, 41BB, and ICOS expression relative to anti-CD3ε activated CD8+ T cells (FIG. 63).

The TEMRA like phenotype of anti-TCR Vβ antibody expanded T cells was further analyzed using time lapse flow cytometry to evaluate expression of CD45RA and CCR7 at different time points post activation. Isolated human T-cells were activated with immobilized (plate-coated) anti-CD3ε or anti-TCR Vβ at 100 nM for between 1-8-days. After each (1, 2, 3, 4, 5, 6, 8-) day activation, T-cell subsets were identified by FACS staining for surface markers for Naïve/TSCM T cell (CD4+/CD8+, CD45RA+, CCR7+), T central memory (TCM; CD4+/CD8+, CD95+, CD45RA−, CCR7+), T effector memory (TEM; CD4+/CD8+, CD95+, CD45RA−, CCR7−), and T effector memory re-expressing CD45RA (TEMRA; CD4+/CD8+, CD95+, CD45RA+, CCR7−). TCRβV+ T-cells are identified by TCR Vβ+ staining. FACS stained samples were analyzed by flow cytometry analysis. Data shown a representative for CD4+ T-cells from 1 of 3 donors.

FIG. 64 shows a series of FACS plots showing the percentage of CD3+ (CD4 gated) TCRβV 6-5+ T cells 1, 2, 3, 4, 5, 6, and 8 days port activation with BCMA and the anti-TCR antibody anti-TCR Vβ 6-5 v1. Analysis of the percentage of CD4+ T cells expanded using isotype control (IgG1 N297A), anti-TCRβV (anti-TCR Vβ 6-5 v1), or anti-CD3ε (OKT3) antibodies on day 0 post activation (FIG. 65A), day 1 post activation (FIG. 65B), day 2 post activation (FIG. 65C), day 3 post activation (FIG. 65D), day 4 post activation (FIG. 65E), day 5 post activation (FIG. 65F), day 6 post activation (FIG. 65G), and day 8 post activation (FIG. 65H). The percentage of TEMRA like T cells expressing both CD45RA and CCR7 shows an increase in the population of TEMRA like cells in the CD4+ TCR Vβ 6-5+ T cell cultures expanded with the anti-TCR Vβ 6-5 v1 antibody compared to those expanded with the OKT3 antibody. Similar results were seen with CD8+ T cells. The results further show that purified human T-cells activated by anti-TCRβV 6-5 directly differentiates to TEMRA subsets and proliferate when compared to purified T-cells activated by anti-CD3ε (OKT3).

In summary, the data shows anti-TCRβV antibodies activated and expanded T cells represent a novel subset of recently activated effector T cells which share phenotypic markers with TEMRA. This is in contrast to anti-CD3e-expanded T cells which differentiated into TCM and TEM. TCRβV expanded T cells are highly proliferative and do not upregulate the senescent marker CD57 OX40, 4-1BB, and ICOS are upregulated on anti-TCRβV activated T cells.

Example 29: Metabolic State of αTCRβV Activated T Cells

To evaluate the metabolic phenotype of T cells activated with αTCRβV antibodies—naïve T cells from PBMCs were stimulated and expanded for 5 days with plate-bound anti-CD3 antibody (OKT3) or anti-TCRβV antibody (anti-TCRβV 6-5 v1 antibody). Activated T cells were then rested in IL-2 containing media for 2 days, before they were cryopreserved. Prior to assay setup, cells were thawed and re-stimulated for 3 days with plate-bound anti-CD3 Ab (clone OKT3) or anti-TCRβV antibody (anti-TCRβV 6-5 v1 antibody), respectively. Equal numbers of live cells were plated on a Seahorse cartridge, and the Real-Time ATP Rate Assay was performed according to manufacturer's instructions. The data showed that ATP production from glycolysis (FIG. 66A) oxidative phosphorylation (FIG. 66B) in T cells from 3 donors (representative results from a single donor presented in FIG. 66A-66B) activated with the anti-TCRβV 6-5 v1 antibody increased compared to T cells activated with the OKT3 antibody (3-fold increase in ATP production was observed on average); and one donor showed equal levels of ATP production in anti-TCRβV 6-5 v1 and OKT3 Ab stimulated cells (data not shown).

The increased mitochondrial respiration in T cells activated with anti-TCRβV 6-5 v1 antibody compared to T cells activated with the OKT3 antibody is further shown in FIG. 67, which shows the oxygen consumption rate (OCR) of T cells from about 0 to 75 minutes activated with the indicated antibody. Data in FIG. 66 is from a single donor; a second donor tested showed equal levels of ATP production in anti-TCRβV 6-5 v1 and OKT3 Ab stimulated cells (data not shown). FIGS. 68A-68C show the oxygen consumption rate (OCR) of T cells activated with the indicated antibody during basal respiration (FIG. 68A), maximal respiration (FIG. 68B), and spare respiratory capacity (FIG. 68C). Cells were plated in media containing glucose and glutamine to measure basal OCR. FCCP (ETC accelerator) was added to the cell culture medium to determine maximum respiratory capacity/max OCR. Antimycin A & Rotenone (ETC inhibitor) were added to the cell culture medium to determine spare respiratory capacity and non-mitochondrial oxygen consumption. The data presented in FIGS. 68A-68C α-TCRβV 6-5 v1 activated T cells had significantly increased basal respiration, maximal respiration, and spare respiratory capacity compared to α-CD3 (OKT3) activated T cells (data from a single donor). A second donor was tested which showed equal levels of ATP production in anti-TCRβV 6-5 v1 and OKT3 Ab stimulated cells (data not shown).FIG. 68D indicates the areas of basal respiration and maximal respiration as shown in FIG. 67A and FIG. 67B, respectively.

In order to determine if the observed increase in metabolism due to differences in T cell stimulation, or is it intrinsic to the differentiation stage of T cells activated with anti-TCRβV antibodies TCRβV 6-5+ T cells were expanded for 5 days with plate-bound anti-TCRβV 6-5 v1 Ab. Cells were then rested in IL-2 containing media for 2 days and cryopreserved. Upon thawing, cells were re-stimulated with anti-TCRβV 6-5 v1 for 3 days. Cells were then counted and equal numbers of live cells were re-seeded and stimulated with plate-bound anti-CD3 Ab (clone OKT3) or anti-TCRβV 6-5 v1, respectively, for 24 hours. Equal numbers of live cells were plated on the Seahorse cartridge and the Real-Time ATP Rate Assay was performed.

The results show that ATP production by glycolysis (FIG. 69A) and oxidative phosphorylation (FIG. 69B) by T cells activated with anti-TCRβV 6-5 v1 is significantly increased upon re-stimulation with α-CD3 antibody OKT3 versus α-TCRβV 6-5 v1 antibody. The observed increase in metabolism of T cells activated with anti-TCRβV 6-5 v1 appears to be due to intrinsic differences upon differentiation into these cells. T cells activated with anti-TCRβV 6-5 v1 have an increased metabolism compared to CD3-activated T cells, which can be further enhanced with strong T cell stimulation via OKT3.

In summary, the results show that T cells activated with anti-TCRβV antibodies have a metabolic memory phenotype. The cells are not metabolically exhausted, because exhausted T cells have a decreased metabolism. α-TCRβV 6-5 v1-stimulation induces a T cell differentiation stage, which is highly metabolically active, indicative of an effector memory phenotype. This metabolic phenotype is maintained when these cells are re-stimulated with other T cell engagers (OKT3).

Example 30: Assessment of CRS with Anti-CD3e Antibody Compared to Anti-TCRβV Antibody

To determine CRS effect of a low affinity (Teneobio) anti-CD3e antibody, a cytokine release assay (CRA) with PBMCs was used. Briefly, PBMCs from two donors were stimulated with plate-coated antibodies: anti-TCRvb6-5 v2, anti-CD3e(SP34) or Teneobio's anti-CD3e antibody. T cell activating antibodies were tested at 100 nM, the highest concentration previously shown not to induce CRS cytokines in this assay. Supernatants were collected at day 1, 3, 5 and 7. Cytokine secretion measurement (IFN-g, IL-10, IL-15, IL-17A, IL-1α, IL-1b, IL-2, IL-4, IL-6 and TNF-α) was detected using MSD analysis. The data show results from two donors.

FIGS. 70A-70F show that a reduced affinity anti CD3e antibody (TeneoBio) induces expression of IFNg, TNFα, IL-1α, IL-1b, IL-6 (CRS and neurotoxicity associated cytokines) similar to the SP34-2 anti CD3e antibody. In contrast, the anti-TCRvb6-5 v2 of the present invention does not induce CRS or neurotoxicity associated cytokines.

In summary the data shows that Tenebio's anti-CD3e antibody induces cytokines associated with CRS and neurotoxicty in this highly sensitive PBMC CRA. Thus, Tenebio's anti-CD3e antibody has potential to induce CRS and NT as seen with SP34-based T cell-redirecting bispecific molecules. The anti-TCRvb6-5 disclosed herein does not induce CRS- and NT-associated cytokines in this assay, suggesting that in some embodiments, TCRvb6-5 based antibodies may be amenable to administration at higher doses and avoid MABEL (minimum anticipated biologic effect level) dosing regimen required for current CD3e-based bispecific molecules.

Example 31: Anti-TCRβV Stimulated PBMC Mediated Stimulation of NK Cell Expansion

To assess whether anti-TCRβV stimulated PBMCs mediate expansion of NK cells in vitro—human PBMCs were stimulated with 100 nM of plate-coated anti-TCRβV 6-5 v1 anti-CD3ε (OKT3 and SP34-2) for up to 7 days. NK cells were identified via FACS staining for CD3-/CD56+/CD16+/NKp46+ populations. NK cell count was determined by a constant μl sample (presented as relative count for each donor). NK cell-mediated target cell lysis was determined 6-days post stimulation, in which PBMCs were harvested and co-cultured with K562 target cells for 4 hours to determine cell killing, via DRAQ7 viability FACS staining.

The results show that anti-TCRβV stimulation increases NK cell numbers compared to OKT3 stimulation (FIG. 71; FIG. 72). FACS CFSE staining further shows NK cell proliferation (FIG. 73). FIG. 74 and FIG. 75 show NK cell mediated lysis of target K562 cells. In summary, anti-TCRβV 6-5 antibody induces expansion of NK cells in PBMC; and this effect is unlikely to be mediated through the FcR on NK cells as anti-CD3ε antibodies did not expand NK cells. Expanded NK cells by anti-TCRβV 6-5 v1 mediate potent target cell (K562) lysis in vitro.

In addition to the experiments conducted above using the anti-TCRβV 6-5 v1 antibody, similar experiments were carried out using anti-TCRβV antibodies that recognize different clonotypes. In one experiment, the anti-TCRβV 12 antibodies: anti-TCRvβ 12-3/4 v1, anti-TCRvβ12-3/4 v2, and anti-TCRvβ 12-3/4 v3 were used to activate/expand PBMCs using solid-phase stimulated (plate-coated) with the indicated T cell-activating antibody at 100 nM for 6 days as described above. Flow analysis was performed for NK cells using NKp46 and CD56 (CD3 negative). Data was generated from 3 donors and representative of 1 independent experiments.

Activation/expansion of the PBMCs with isotype control or the anti-CD3ε antibody OKT3 or SP34-2 did not induce expansion of NK cells (FIG. 76; FIG. 78). However, activation/expansion of PBMCs with anti-TCRvβ 12-3/4 v1 (FIG. 77), anti-TCRvβ 12-3/4 v2 (FIG. 77), and anti-TCRvβ 12-3/4 v3 (FIG. 78) all induced NK cell expansion. In summary, the data shows that anti-TCRvb 12 antibodies are able to induce indirect expansion of NK cells from PBMC cultures in vitro.

Example 32: Concentration Response to Anti-TCRβV Stimulation In Vitro

Human PBMCs were solid-phase stimulated (plate-coated) with the indicated T cell-activating antibody at the indicated different concentrations: i) anti-TCRvb 6-5 v1 antibody; ii) OKT3 (anti-CD3ε antibody); or iii) SP34-2 (anti-CD3ε antibody). Supernatant were collected on day 1, day 3 and day 5 and cytokines quantified by using Meso Scale Discovery (MSD) assay. The production of cytokines IFNγ (FIG. 79), IL-2 (FIG. 80), IL-15 (FIG. 81), IL-1β (FIG. 82), IL-6 (FIG. 83), and IL-10 (FIG. 84) was analyzed. The results indicate that the lack of CRS associated cytokine induction by T cells activated with an anti-TCRvb is not a result of inhibition or toxicity due to high antibody concentrations.

Example 33: T Cells Activated by Anti-TCRβV Antibodies have a Distinct Cytokine Release Profile Compared to T Cell Activated with Anti-CD3ε Antibodies

To assess the cytokine release profile of T cells activated/expanded using anti-TCRβV antibodies as compared to anti-CD3ε antibodies—PBMCs were cultured in cell culture plates coated with the immobilized anti-TCRβV antibody anti-TCRβV 6-5 v1 or an anti-CD3ε antibody, either OKT3 or SP37-2. The cells were cultured for 1-8 days, the supernatant collected, and cytokines analyzed using Meso Scale Discovery (MSD) assay. T cells samples from numerous different human donors were tested.

FIG. 85 shows a summary of data from 17 donors. The highest overall cytokine secretion from time points (day 3 and beyond) was used for further analysis. Each data point was normalized against the highest secretion for each donor and showed as relative % of highest (at a confidence interval of 0.95 percentile). The data shows that T cells activated/expanded with an anti-TCRβV antibody as compared to anti-CD3ε antibody release less IFNγ, TNFα, IL-1β, IL-4, IL-6, IL10, and IL-17; while releasing an increased amount of IL-2 (FIG. 85).

A series of experiments using the methods previously described, but varying the culture period were conducted with PBMCs from different donors. In one experiment, PBMCs from four different donors were cultured in plates coated with immobilized anti-TCRβV antibody anti-TCRβV 6-5 v1 or an anti-CD3ε antibody, either OKT3 or SP37-2 for 1-6 days. The data confirms that T cells activated/expanded with an anti-TCRβV antibody as compared to anti-CD3ε antibody release lower levels of IFNγ (FIG. 86A), IL-1β (FIG. 86B), IL-4 (FIG. 86C), IL-6 (FIG. 86D), IL10 (FIG. 86E), and TNFα (FIG. 86F); and higher levels of IL-2 (FIG. 86G).

In a second experiment, PBMCs from six different donors were cultured in plates coated with immobilized anti-TCRβV antibody, either anti-TCRβV 6-5 v1 or anti-TCRβV 6-5 v1; or an anti-CD3ε antibody, either OKT3 or SP37-2 for 1-6 days, or isotype control. The data confirms that T cells activated/expanded with an anti-TCRβV antibody as compared to anti-CD3ε antibody release lower levels of IFNγ (FIG. 87A), IL-1β (FIG. 87B), IL-4 (FIG. 87C), IL-6 (FIG. 87D), IL10 (FIG. 87E), and TNFα (FIG. 87F); and higher levels of IL-2 (FIG. 87G).

In a third experiments, PBMCs from three different donors were cultured in plates coated with immobilized anti-TCRβV antibody, either anti-TCRβV 6-5 v1 or anti-TCRβV 6-5 v1; or an anti-CD3ε antibody, either OKT3 or SP37-2 for 1-8 days, or isotype control. The data confirms that T cells activated/expanded with an anti-TCRβV antibody as compared to anti-CD3ε antibody release lower levels of IFNγ (FIG. 88A), IL-1β (FIG. 88B), IL-4 (FIG. 88C), IL-6 (FIG. 88D), IL10 (FIG. 88E), and TNFα (FIG. 88F); and higher levels of IL-2 (FIG. 88G).

In a fourth experiments, PBMCs from two different donors were cultured in plates coated with immobilized anti-TCRβV antibody, either anti-TCRβV 6-5 v1 or anti-TCRβV 6-5 v1; or an anti-CD3ε antibody, either OKT3 or SP37-2 for 2-7 days, or isotype control. The data confirms that T cells activated/expanded with an anti-TCRβV antibody as compared to anti-CD3ε antibody release lower levels of IL-17A (FIG. 89A). In a fifth experiments, PBMCs from four different donors were cultured in plates coated with immobilized anti-TCRβV antibody, either anti-TCRβV 6-5 v1 or anti-TCRβV 6-5 v1; or an anti-CD3ε antibody, either OKT3 or SP37-2 for 2-8 days, or isotype control. The data confirms that T cells activated/expanded with an anti-TCRβV antibody as compared to anti-CD3E antibody release lower levels of IL-17A (FIG. 89B). In a sixth experiments, PBMCs from two different donors were cultured in plates coated with immobilized anti-TCRβV antibody, either anti-TCRβV 6-5 v1 or anti-TCRβV 6-5 v1; or an anti-CD3ε antibody, either OKT3 or SP37-2 for 2-7 days, or isotype control. The data confirms that T cells activated/expanded with an anti-TCRβV antibody as compared to anti-CD3ε antibody release lower levels of IL-17A (FIG. 89C). In a seventh experiments, PBMCs from two different donors were cultured in plates coated with immobilized anti-TCRβV antibody, either anti-TCRβV 6-5 v1 or anti-TCRβV 6-5 v1; or an anti-CD3ε antibody, either OKT3 or SP37-2 for 2-7 days, or isotype control. The data confirms that T cells activated/expanded with an anti-TCRβV antibody as compared to anti-CD3ε antibody release lower levels of IL-17A (FIG. 89D).

A series of similar experiments were conducted using the TCRβV antibody anti-TCRβV 6-5 v1 or anti-TCRvb 12-3/4 v1 to further assess the cytokine release profile of T cells activated/expanded using anti-TCRβV antibodies as compared to anti-CD3ε antibodies. As described above, PBMCs were cultured in cell culture plates coated with the immobilized anti-TCRβV antibody, anti-TCRβV 6-5 v1 or anti-TCRvb 12-3/4 v1; or an anti-CD3ε antibody, either OKT3 or SP37-2; isotype control; or anti-TCRβV 6-5 v1 in combination with. The cells were cultured for 1-8 days, the supernatant collected, and cytokines analyzed using Meso Scale Discovery (MSD) assay. Data generated from 2 donors and representative of 2 independent experiments.

The data confirmed that T cells activated/expanded by either anti-TCRβV antibody, anti-TCRβV 6-5 v1 or anti-TCRvb 12-3/4 v1, as compared to either anti-CD3ε antibody (OKT3 or SP37-2) secreted a lower level of IFNγ (FIG. 90A), IL-1β (FIG. 90B), IL-4 (FIG. 90C), IL-6 (FIG. 90D), IL10 (FIG. 90E), TNFα (FIG. 90F); and higher levels of IL-2 (FIG. 90G). Secretion of IL-12p70 (FIG. 9011), IL-13 (FIG. 901), IL-8 (FIG. 90J), Exotaxin (FIG. 90K), Exotaxin-3 (FIG. 90L), IL-8 (FIG. 90M), IP-10 (FIG. 90N), MCP-1 (FIG. 90O), MCP-4 (FIG. 90P), MDC (FIG. 90Q), MIP-1α(FIG. 90R), MIP-1b (FIG. 90S), TARC (FIG. 90T), GMCSF (FIG. 90U), IL-12-23p40 (FIG. 90V), IL-15 (FIG. 90W), IL-16 (FIG. 90X), IL-17a (FIG. 90Y), IL-1a (FIG. 90Z), IL-5 (FIG. 90AA), IL-7 (FIG. 90BB), TNF-β (FIG. 90CC), and VEGF (FIG. 90DD), were also tested.

In addition to determining the cytokine profile of T cells activated with the αTCRβV antibodies αTCRβV 6-5 v1 and αTCRβV 6-5 v2 (described above); the assays were conducted with additional αTCRβV antibodies recognizing different clonotypes.

In one series of experiments antibodies tested included anti-TCRvb 12-3/4 v1, anti-TCRvb 10, and anti-TCRvb 5. Per the protocol described above, human PBMCs were solid-phase stimulated (plate-coated) with the indicated T cell-activating antibody (anti-TCRvb 12-3/4 v1, anti-TCRvb 10, anti-TCRvb 5, or the anti-CD3ε antibody SP34) at 100 nM. Supernatant were collected on day 1 to day 8; and cytokines were quantified using Meso Scale Discovery (MSD) assay. 91 provides a graphical representation of sequences between the different clonotypes, highlighting the four subfamilies tested in this series of experiments. PBMCs activated/expanded with the anti-TCRvb 12-3/4 v1 antibody (FIG. 92A), anti-TCRvb 10 antibody (FIG. 92B), or anti-TCRvb antibody (FIG. 92C) exhibited lower levels of secretion of cytokines associated with cytokine release syndrome, including IFNγ, TNFα, IL-1β, IL-2, IL-6, and IL-10, as compared to PBMCs activated/expanded with the anti-CD3ε antibody SP34-2.

In a second series of experiments, antibodies tested included the anti-TCRVβ antibodies: BJ1460, BJ1461, BJ1465, BJ1187, BJM1709; the anti-CD3ε antibody OKT3, and a cell only control. At Day-0 PBMCs from donor 10749 were thawed and counted along with PBMCs from two fresh donors (13836 and 14828). 200,000 PBMCs in 180 uL of X-vivo media/well (1×10e6 cells/mL) was added to a round bottom 96 well plate—one donor for ⅓ of the plate. 20 uL of 10× TCRVβ antibodies at 100 nM or 15 μg/mL were added to the wells of the plate and one triplicate of wells was added with cells only. The pate was kept in a 37° C. incubator with 5% CO2. The cells were stimulated for 3 days with a selected antibody and 504, of supernatant harvested from the plate and stored at −20° C. 504, of media was added back to each well and the plate kept in a 37° C. incubator with 5% CO2. On Day-6 50 uL of supernatant was harvested from each well of the plate and stored at −20° C. The cells from two wells out of the triplicate were combined and media replenished with huIL-2 was added the cell suspension for each donor was transferred into a 12-well plate. The cells were incubated overnight to allow for rest and expansion in IL-2. The cells were subsequently stained for specific VP-clones for detection of specific VP-clone expansion by FACS analysis. The concentration of cytokines (including IFNγ, IL-10, IL-17A, IL-1α, IL-1β, IL-2, IL-6, and TNFα) in the media were analyzed in the Day-3 and Day-6 supernatant samples using Meso Scale Discovery (MSD) assay. The data confirmed that PBMCs cells activated/expanded using any of the anti-TCRβV antibodies—BJ1460, BJ1461, BJ1465, BJ1187, BJM1709—secreted lower levels of IFNγ (FIG. 93A), IL-10 (FIG. 93B), IL-17A (FIG. 93C), IL-1α (FIG. 93D), IL-1β (FIG. 93E), IL-6 (FIG. 93F), TNFα (FIG. 93G); and higher levels of IL-2 (FIG. 9311). FACS analysis further showed expansion of T cells expressing the indicated TCRVβ clones (FIG. 94).

In a third series of experiments, antibodies tested included the anti-TCRVβ antibodies: BHM1675, BJM0816, BJ1188, BJ1189, BJ1190; and the anti-CD3ε antibody SP34-2. The indicated antibodies were coated into a 96-well round bottom plate at concentration of 100 nM or 15 μg/mL at 200 μl/well in PBS at 4° C. overnight or at 37° C. for a minimum of 2 hours. The plate was washed the next day with 2004, of PBS and 0.2×10{circumflex over ( )}6 PBMCs/well from donors: CTL_123, CTL_323 and CTL_392. Supernatant samples were collected on days 1, 3, 5, and 7. A 10-plex Meso Scale Discovery (MSD) assay was run on the supernatants to determine the concentration of cytokines (including IFNγ, IL-10, IL-17A, IL-1α, IL-1β, IL-6, IL-4, and IL-2). After day 7, cells were pelleted and added to culture medium supplemented with IL-2 for one additional day to allow for expansion. Expansion of T cells expressing TCRVβ clones was analyzed by FACS staining using the same activating antibody followed by a secondary anti-human/mouse FITC antibody. Live/Dead, CD4+ and CD8+ T cells were also stained for using BHM1675, BJM0816, BJ1189 and BJ1190 antibodies. The data confirmed that PBMCs cells activated/expanded using any of the anti-TCRβV antibodies—BHM1675, BJM0816, BJ1188, BJ1189, BJ1190—secreted lower levels of IFNγ (FIG. 95A), IL-10 (FIG. 95B), IL-17A (FIG. 95C), IL-1α (FIG. 95D), IL-10 (FIG. 95E), IL-6 (FIG. 95F), IL-4 (FIG. 95G); and higher levels of IL-2 (FIG. 9511). FACS analysis further showed that TCRVβ sub-clone T-cells are expanded by their respective activation antibody (FIG. 96).

In a fourth series of experiments, antibodies tested included the anti-TCRVβ antibodies: BJ1538, BJ1539, BJ1558, BJ1559, BHM1709; and the anti-CD3ε antibody OKT3. The indicated antibodies were coated into a 96-well round bottom plate at concentration of 100 nM or 15 μg/mL at 200 μl/well in PBS at 4° C. overnight or at 37° C. for a minimum of 2 hours. The plate was washed the next day with 200 μL of PBS and 0.2×10{circumflex over ( )}6 PBMCs/well from donors: 10749, 5078 and 15562 (frozen and thawed samples). Supernatant samples were collected on days 3 and 6. A 10-plex Meso Scale Discovery (MSD) assay was run on the supernatants to determine the concentration of cytokines (including IFNγ, IL-10, IL-17A, IL-1α, IL-1β, IL-6, IL-4, TNFα, and IL-2). The data confirmed that PBMCs cells activated/expanded using any of the anti-TCRβV antibodies—BJ1538, BJ1539, BJ1558, BJ1559, BHM1709—secreted lower levels of IFNγ (FIG. 97A), IL-10 (FIG. 97B), IL-17A (FIG. 97C), IL-1α (FIG. 97D), IL-10 (FIG. 97E), IL-6 (FIG. 97F), IL-4 (FIG. 97G) TNFα (FIG. 97I1); and higher levels of IL-2 (FIG. 97I).

In summary, the data shows that anti-TCRvb antibodies recognizing different TCRvb subfamilies (or subtypes) have a similar cytokine profile and do not induce cytokines associated with CRS.

Example 34: Anti-TCRvb does not Activate T Cells without Cross-Linking

To assess whether bivalent anti-TCRvb antibodies activate T cells without cross-linking—purified T cells from 2 donors were stimulated with anti-TCRvb (TCRvb 6-5 v1) or anti-CD3e (SP34), either plate-coated or in solution. Supernatants were collected at day 1, 3, 5 and 7 post activation. Cytokine secretion was detected using MSD 10 plex kit (IFN-g, IL-10, IL-15, IL-17A, IL-1α, IL-1b, IL-2, IL-4, IL-6 and TNF-α).

The results show the PBMCs activated/expanded with anti-TCRvb 6-5 v1 antibody in solution do no induce very little IFNγ secretion as compared to PBMCs activated/expanded with anti-TCRvb 6-5 v1 antibody in immobilized (allowing for crosslinking) (FIG. 98A and FIG. 98B). The results show the PBMCs activated/expanded with anti-TCRvb 6-5 v1 antibody in solution do no induce very little or no IL-1b (FIG. 98C), IL-10 (FIG. 98E), IL-15 (FIG. 98F), IL-17A (FIG. 98G), IL-1a (FIG. 98H), IL-1b (FIG. 98I), IL-2 (FIG. 98J), IL-4 (FIG. 98K), IL-6 (FIG. 98D), and TNF-α (FIG. 98L) secretion. In summary, the data shows that anti-CD3s activates T cells in solution (without crosslinking); while the anti-TCRvb antibodies does not activate T-cells in solution.

Example 35: Competition of Binding to TCRVB by Two Anti-TCRVβ 5-5,5-6 Antibodies with Distinct Sequences

This Example describes epitope competition of two anti-TCRVβ 5-5,5-6 antibodies for binding to their shared TCRVB antigen. The TM23 and MH3-2 antibodies both bind to TCRVβ 5-5,5-6. However, the TM23 and MH3-2 antibodies do not share substantial sequence homology. As shown in FIG. 24A-B, anti-TCRβV antibody molecules disclosed herein recognize a structurally conserved domain on the TCRBV protein (as denoted by the circled area in FIG. 24A), but have low sequence similarity among themselves. To test whether two anti-TCRVβ 5-5,5-6 antibodies which do not share substantial sequence homology, can compete for binding to the TCRBV antigen, a competition assay was carried out.

Purified MH3-2 antibody was conjugated to AF647. T cells from two donors were preincubated with 500 nM of the TM23 antibody or left untreated. The T cells were then stained with the MH3-2 antibody conjugated to AF647.

The results show that preincubation of T cells with the TM23 antibody blocks binding of MH3-2 (FIG. 99 and FIG. 100). The data shows that the TM23 antibody competes for binding with the same epitope as the MH3-2 antibody, despite both antibodies having diverse sequences. This data confirms the observation that anti-TCRβV antibody molecules which have low sequence similarities among themselves, bind and recognize a structurally conserved epitope on the TCRBV protein.

Example 36. Polyfunctional Strength Index of Anti-TCRVβ6-5 Antibody Expanded T Cells

The polyfunctional strength index (PSI) of PBMCs were compared to anti-CD3E antibody expanded CD4+ T cell (FIG. 101A) and CD8+ T cells (FIG. 101B) and anti-TCRVβ 6-5 antibody expanded (Drug Expanded T cells) CD4+ T cells (FIG. 101A) and CD8+ T cells (FIG. 101B). PSI is defined as the percentage of polyfunctional cells in the sample, multiplied by the intensities of the secreted cytokines. The data shows that there is a greater upregulation of PSI in the CD4+ T cells (FIG. 101A) and CD8+ T cells (FIG. 101B) across the groups expanded with anti-TCRVβ6-5 antibody.

Example 37: Binding of TCRVB/CD19 Bispecific to Soluble TCR and Jurkat Cells Expressing TCR

In this Example, the binding affinity of a TCRVB/CD19 bispecific (FIG. 102A) for the TCR was tested.

Jurkat cells expressing TRBV6-5 were stained with increasing concentrations of bispecific molecule CD19×TCRvb6-5 (2×2) or control antibody TCRvb6-5 v1 at 4 Celsius for 30 min. Subsequently, the cells were washed with PBS buffer and antibodies bound to the surface of the cells detected by PE-labeled anti-human Fc antibody. The percentage of the positive stained cells were blotted against the concentration. FIG. 102B shows the binding of a TCRVB/CD19 bispecific to soluble TCR. The bispecific molecule binds soluble TCR with a Kd of 12 nM.

Bispecific CD19×TCRvb6-5 (2×2) antibody was immobilized on a CM5 Series S Sensor Chip via Anti-human Fc antibody to 50 RU. Soluble TRBV6-5 antigen was diluted to 500 nM and then serially diluted two-fold. Association was 180 seconds, dissociation was 300 seconds. This assay was run in 1×HBS-EP+ Buffer pH 7.4 and at 25C. The data was fit using a 1:1 binding model. FIG. 102C shows the binding of a TCRVB/CD19 bispecific or the anti-TCRVB 6-5 v1 antibody to TCR expressed on Jurkat cells. The EC50 for the anti-TCRVB 6-5 v1 antibody was 0.6486 and the EC50 for the bispecific molecule was 1.720.

Example 38: In Vitro and In Vivo Characterization of a Murine Anti-TCRVB Antibody

This Examples describes the characterization of a murine anti-TCRVB antibody. Similar to the human clonotypes (subfamilies), the TCRb variable chain locus in mice consists of 31 different families with a total of 35 subfamilies of which 23 are functionally expressed. A surrogate TCRvb clonotypical antibody for mice strain C57BL/6 has been identified which shares similar characteristics with the human TCRvb antibodies. This anti-mouse TCRvb antibody binds specifically to TCRvb 13-2 and 13-3 in C57BL/6 mice which are expressed on approximately 15% of all T cells. Similar to the human TCRvb specific antibodies, this murine TCRvb specific antibody (TCRvb 13-2/3) induces murine T cell proliferation and a similar cytokine profile in vitro. The discovery of TCRvb 13-2/3 will enable the evaluation of TCRvb-mediated T cell activation and re-directed cell killing in fully immuno-competent mice models, as well as to assess memory anti-tumor response in vivo.

FIG. 103A shows the schematic of a bispecific molecule which was used in the following experiments. The bispecific molecule recognizes murine CD19 and murine TCRVB 13-2, 13-3 (also referred to as mCD19×mTCRvb 13-2/3). The bispecific molecule did not bind to mouse Fc gamma receptors.

First, the in vitro functional activity of the murine bispecific molecule was tested. Splenic mononuclear cells were freshly isolated from C57BL6 mice, treated with mCD19×mTCRvb 13-2/3 (2×2). The isolated cells were assessed for B cell depletion, TCRvβ+ T cell binding, expansion and activation. Cells were treated with 0.0008-200 nM doses (4-fold dilutions) of mCD19×mTCRvb 13-2/3 (2×2) in RPMI-1640 with 10% FBS, or medium alone, for 6 days. On day 3 and 6, cells were analyzed by flow cytometry using the antibodies shown below:

TABLE 23 Antibodies Primary Ab Secondary Ab eFLuor780 Viability CD20 A647 CD3 BV421 TCRVβ 13-2/3 PE CD25 FITC

FIG. 103B shows that the mCD19×mTCRvb 13-2/3 (2×2) bispecific antibody bound specifically to splenic T cells from C57BL6 mice. Activation and expansion of mTCRvβ+ T cells was observed at day 3 and 6, respectively (FIG. 103C). Following 6 days of treatment, mCD19×mTCRvb 13-2/3 (2×2) induced efficient murine B cell depletion in C57BL6 splenocytes in vitro (FIG. 103D). Taken together, the in vitro characterization shows that mCD19×mTCRvb 13-2/3 (2×2) could serve as a surrogate tool for syngeneic tumor model studies.

Next, in vivo experiments were performed with the murine bispecific molecule. On day 0, 8 week old female C57BL/6 mice were randomized in to three arms (n=5/arm) based on body weight. Mice were intravenously injected once either with PBS, 0.1 mg/kg and 1 mg/kg mCD19×mTCRvb 13-2/3 (2×2). On Day 3 mice were sacrificed and harvested for whole blood and spleen. Tissues were subjected to Flow cytometry and checked for B-cells, NK cells, TCRvb+ cells and CD3+ cells.

The results show that the murine mCD19×mTCRvb 13-2/3 bispecific molecule depletes B cells in the blood and spleen of animals (FIG. 104). The murine bispecific molecule also expands mouse NK cells in vivo, in the blood and spleen (FIG. 105). The study also demonstrated that mCD19×mTCRvb 13-2/3 (2×2) is well tolerated at the indicated doses and duration of study.

Example 39: Target Cell Lysis and Cytokine Profile of CD19×TCRvβ Bispecific Molecule

This Example describes potent lysis of target cells and reduced CRS associated cytokine secretion with a CD19×TCRvβ bispecific molecule.

To test target cell killing, αTCRvβ 6-5 v1 pre-expanded T cells were incubated with Raji target cells in the presence of an CD19×TCRvβ bispecific molecule, CD19×CD3 bispecific molecule or αTCRvβ 6-5 v1 antibody (non-targeted) for 24 hours. Target cell lysis was assessed by a KILR Cytotoxicity and Cytokine Quantification as follows. Human PBMCs were isolated from whole blood. From isolated PBMC's, human CD3+ T cells were isolated using magnetic-bead separation (negative selection) (Miltenyi biotec) and activated by immobilized (plate-coated) anti-TCR V013.1 (BHM1709) at 100 nM for 6 days. Activated T-cells (from plate-coated) were then transferred and expanded in tissue culture flask in the presence of human IL-2 at a concentration of 50 U/ml for an additional 2 days. Expanded TCR V013.1 were washed and co-cultured in the presence of CD19-expressing Raji Cells (target cells) at a E:T ratio of 5:1 and serial diluted concentration of T-cell engager bispecific antibodies including, anti-TCR V013.1/CD19 (BJM0093), anti-CD3/CD19 (BJM0030) and anti-TCR V013.1 (BHM1709, serving as control) for 24 hours. Post 24 hours, cell co-culture supernatants were collected and quantified for specific target cell death. Target cells (Raji cells) is a KILR-retroparticles reporter cell assay (DiscoverX).

KILR-Raji Target cells are engineered to stably express a protein tagged with enhanced ProLabel (ePL), a 0-gal reporter fragment, using the KILR Retroparticles, and when its membrane is compromised due to cell death, it will release the tagged protein into the media. The KILR reporter protein is detected in the media/supernatant by the addition of detection reagents containing the enzyme acceptor (EA) fragment of the 0-gal reporter. This leads to the formation of the active β-gal enzyme which hydrolyzes the substrate to give a chemiluminescent output (RLU). Percentage (%) of target cell death is calculated using the following formula: (RLU Treatment—RLU No Treatment). FIG. 106B shows the results of this assay. αTCRvβ 6-5 v1 pre-expanded T cells (TrEKs) demonstrate efficient killing of Raji target cell at low effector to target ratios of 0.25:1.

Next, B cell depletion with the CD19×TCRvβ bispecific molecule was tested. Purified T-cells and purified B-cells from the same donor were treated with anti-TCRvβ/CD19 bispecific or Amgen's Blincyto for 2-6 days. B-cell depletion was measured by anti-CD20 staining via FACS analysis. As shown in FIG. 106C-D, CD19×TCRvβ 6-5 (2×2) bispecific concept molecule induces B cell depletion 6 days after co-incubation with target cells. A similar result was observed when PBMC's were treated with the anti-TCRvβ/CD19 bispecific or anti-CD3/CD19 bispecific for 1-6 days followed by measurement of B-cell depletion by anti-CD20 staining via FACS analysis (FIG. 106D-E). This data shows that the anti-TCRvβ/CD19 bispecific requires time for differentiation and expansion of TCRvβ+ T cells.

To determine if the lack of CRS associated cytokine induction by immobilized anti-TCR Vb antibodies can be recapitulated by the bispecific molecule targeting CD19, human PBMCs were incubated in the presence of T cell-activating antibody bispecific molecules at 3 nM. Compounds tested/compared were: CD19×TCRvb and CD19×CD3e. Supernatant were collected on day 1 to day 6, and cytokines were quantified by using MSD.

FIG. 107A-B show that the CD19×TCRvβ (2×2) bispecific molecule shows increased IL-2 production against a backdrop of delayed and reduced levels of CRS-related cytokines compared to the CD19×CD3e bispecific molecule.

Example 40: Pharmacokinetic (PK) Profile of CD19×TCRvβ 6-5 (2×2) in Mice

This Example describes the pharmacokinetic (PK) profile of CD19×TCRβ 6-5 (2×2) in mice to guide the dosing and/or schedule treatment decision for the efficacy study. The study design is shown in FIG. 108A. Briefly, on day 0, 6-8 week old female NSG mice were implanted subcutaneously with 1×10(6) Raji leukemia cells. On day 2, mice were humanized by injecting 10×10(6) human PBMCs via the peritoneal cavity. On day 9, mice were treated with a single dose of CD19×TCRβ 6-5 (2×2) intravenously. Serum was harvested from animals by submandibular bleed at 0, 0.5, 1, 6, 24,48,72, 96, 148 hours (n=3 per time point). Serum drug concentration was measured by Sandwich ELISA.

As shown in FIG. 108B and Table 22 below, the serum half-life of CD19×TCRβ 6-5 (2×2) in tumor bearing humanized NSG animals is approximately 24 hours following a single dose of 1 mg/kg via iv route. This data allowed dose and schedule determination for efficacy studies. Exposures at this dose allow coverage above cellular EC90 beyond 100 hours.

TABLE 22 CD19 × TCRvβ 6-5 PK profile Parameter Value T(½) 23.89 h Tmax 0.5 h Cmax 123 nM AUC0-t 2436.60 mol/*h

Example 41. Optimization of α-TRBV6-5 Antibody

The anti TRBV6-5 antibody was optimized to improve affinity for the human and cyno antigen, improve thermal stability, and remove sequence motifs that might pose chemical stability liabilities. ScFv libraries were built using random mutagenesis (Caldwell et al. (1992) Randomization of genes by PCR mutagenesis. PCR Meth. Appl. 2:28) or a modified version of Kunkel mutagenesis (Kunkel TA. (1985) Rapid and efficient site-specific mutagenesis without phenotypic selection. PNAS 82(2): 488-92). For affinity improvement, library selections vs human and cyno antigens were performed using standard phage display (Lee, C M et al. (2007) Selection of human antibody fragments by phage display. Nature protocols 2, 3001) and yeast display techniques (Chao G, et al. (2006) Isolating and engineering human antibodies using yeast surface display. Nature Protocols. 1(2):755-69). Thermal challenge of phage or yeast populations was used to select for clones with improved thermal stability. Selections were followed by standard screening methods such as ELISA and flow cytometry to identify individual clones with improved properties. Following hit sequencing and analysis of mutation-activity correlation, second-generation libraries were constructed using the same methods above. Library selections and individual clone screening were repeated as above with the modification that more stringent conditions were applied to select for clones with maximized activity. Following hit sequencing, scFv genes were reformatted into the biologically relevant antibody format for expression, purification, and triaging.

Exemplary Embodiments

Disclosed herein are, inter alia, antibody molecules directed to the variable chain of the beta subunit of TCR (TCRβV) which bind and, e.g., activate or expand, T cells, e.g., a subset of T cells (“anti-TCRβV antibody molecules”). In some embodiments, the anti-TCRβV antibody molecules disclosed herein result in a cytokine profile, e.g., a cytokine secretion profile, that differs from that of a T cell engager that binds to a receptor or molecule other than a TCRβV region (“a non-TCRβV-binding T cell engager”). In some embodiments, the anti-TCRβV antibody molecules disclosed herein result in lesser, minimal, or no production of cytokines associated with cytokine release syndrome (CRS), e.g., IL-6, IL-1beta, IL-10 and TNF alpha; and enhanced and/or delayed production of IL-2 and IFN-gamma. In some embodiments, the anti-TCRβV antibodies disclosed herein result in expansion of an immune cell, e.g., a T cell, a tumor infiltrating lymphocyte (TIL), an NK cell, or other immune cells (e.g., as described herein). Also provided herein are methods of making said anti-TCRβV antibody molecules, and methods of using said anti-TCRβV antibody molecules including, methods of using an anti-TCRβV antibody molecule for expanding an immune cell or an immune cell population, and method of using an anti-TCRβV antibody molecule for treating cancer, including the use as combination therapy with TIL and immune checkpoint therapeutics. This disclosure further provides multispecific molecules, e.g., bispecific molecules, comprising said anti-TCRβV antibody molecules. In some embodiments, compositions comprising anti-TCRβV antibody molecules of the present disclosure, can be used, e.g., to activate and/or redirect T cells to promote tumor cell lysis for cancer immunotherapy. In some embodiments, compositions comprising anti-TCRβV antibody molecules as disclosed herein limit the unwanted side-effects of CRS and/or NT, e.g., CRS and/or NT associated with anti-CD3e targeting.

In some embodiments, the anti-TCRβV antibody molecules disclosed herein result in lesser, minimal, or no production of cytokines associated with cytokine release syndrome (CRS), e.g., IL-6, IL-1beta, IL-10 and TNF alpha; and enhanced and/or delayed production of IL-2 and IFN-gamma, compared with an anti-CD3 antibody molecule (e.g., a low affinity anti-CD3 antibody molecule). In some embodiments, administration of the anti-TCRβV antibody molecules disclosed herein in a subject results in reduced cytokine release syndrome (CRS) (e.g., lesser duration of CRS or no CRS), a reduced severity of CRS (e.g., absence of severe CRS, e.g., CRS grade 4 or 5), reduced neurotoxicity (NT), or a reduced severity of NT, compared with similar administration of an anti-CD3 antibody molecule (e.g., a low affinity anti-CD3 antibody molecule).

Accordingly, provided herein are, anti-TCRβV antibody molecules, multispecific or multifunctional molecules (e.g., multispecific or multifunctional antibody molecules) (also referred to herein as a “composition”) that comprise anti-TCRβV antibody molecules, nucleic acids encoding the same, methods of producing the aforesaid molecules, pharmaceutical compositions comprising aforesaid molecules, and methods of treating a disease or disorder, e.g., cancer, using the aforesaid molecules. The antibody molecules and pharmaceutical compositions disclosed herein can be used (alone or in combination with other agents or therapeutic modalities) to treat, prevent and/or diagnose disorders and conditions, e.g., cancer, e.g., as described herein.

In one aspect, the disclosure provides an antibody molecule, e.g., a non-murine, e.g., a human-like (e.g., a human, or humanized antibody molecule), which binds, e.g., specifically binds, to a T cell receptor beta variable (TCRβV) region.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain of an antibody disclosed in any of Tables 1-2, or 10-13, or a sequence with at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity thereto. In some embodiments, the anti-TCRβV antibody molecule comprises a leader sequence comprising the amino acid sequence of SEQ ID NO: 3288. In some embodiments, the anti-TCRβV antibody molecule does not comprise a leader sequence comprising the amino acid sequence of SEQ ID NO: 3288.

In some embodiments, binding of the anti-TCRβV antibody molecule to a TCRβV region results in a cytokine profile, e.g., a cytokine secretion profile, (e.g., comprising one or more cytokines and/or one or more chemokines), that differs from that of a T cell engager that binds to a receptor or molecule other than a TCRβV region (“a non-TCRβV-binding T cell engager”).

In some embodiments, the cytokine profile, e.g., cytokine secretion profile, comprises one, two, three, four, five, six, seven, or all of the following:

    • (i) increased level, e.g., expression level, and/or activity of IL-2;
    • (ii) reduced level, e.g., expression level, and/or activity of IL-1(3;
    • (iii) reduced level, e.g., expression level, and/or activity of IL-6;
    • (iv) reduced level, e.g., expression level, and/or activity of TNFα;
    • (v) reduced level, e.g., expression level, and/or activity of IL-10;
    • (vi) a delay, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more hours delay, in increased level, e.g., expression level, and/or activity of IL-2;
    • (vii) a delay, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 hours delay, in increased level, e.g., expression level, and/or activity of IFN-gamma; or
    • (viii) increased level, e.g., expression level, and/or activity of IL-15, e.g., wherein (i)-(viii) are relative to the cytokine profile, e.g., cytokine secretion profile, of the non-TCRβV-binding T cell engager.

In some embodiments, binding of the anti-TCRβV antibody to a TCRβV region results in reduced cytokine storm, e.g., reduced cytokine release syndrome (CRS) and/or neurotoxicity (NT), as measured by an assay of Example 3, e.g., relative to the cytokine storm induced by the non-TCRβV-binding T cell engager.

In some embodiments, binding of the anti-TCRβV antibody to a TCRβV region results in one, two, three or all of:

    • (ix) reduced T cell proliferation kinetics;
    • (x) cell killing, e.g., target cell killing, e.g. cancer cell killing, e.g., as measured by an assay of Example 4;
    • (xi) increased Natural Killer (NK) cell proliferation, e.g., expansion; or
    • (xii) expansion, e.g., at least about 1.1-10 fold expansion (e.g., at least about 1.1, 1.2, 1.3, 1.4, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, or 10 fold expansion), of a population of T cells having a memory-like phenotype, e.g., as described herein, e.g., wherein (ix)-(xii) are relative to the non-TCRβV-binding T cell engager.

In some embodiments, an anti-TCRβV antibody molecule disclosed herein recognizes (e.g., binds to), a structurally conserved domain on the TCRβV protein (e.g., as denoted by the circled area in FIG. 24A).

In some embodiments, an anti-TCRVβ antibody disclosed herein comprises an Fc region, e.g., as described herein. In some embodiments, the Fc region is a wildtype Fc region, e.g., a wildtype human Fc region. In some embodiments, the Fc region comprises a variant, e.g., an Fc region comprising an addition, substitution, or deletion of at least one amino acid residue in the Fc region which results in, e.g., reduced affinity for and/or binding to, at least one Fc receptor. In some embodiments, the reduced affinity is compared to an otherwise similar antibody with a wildtype Fc region.

In some embodiments, an anti-TCRVβ antibody comprising a variant Fc region has one or more of the following properties: (1) reduced effector function (e.g., reduced ADCC, ADCP and/or CDC); (2) reduced binding to one or more Fc receptors; and/or (3) reduced binding to C1q complement. In some embodiments, the reduction in any one, or all of properties (1)-(3) is compared to an otherwise similar antibody with a wildtype Fc region.

In some embodiments, an anti-TCRVβ antibody comprising a variant Fc region has reduced affinity to a human Fc receptor, e.g., FcγR I, FcγR II and/or FcγR III. In some embodiments, the anti-TCRVβ antibody comprising a variant Fc region comprises a human IgG1 region or a human IgG4 region.

In some embodiments, an anti-TCRVβ antibody disclosed herein comprises any one or all, or any combination of Fc region variants, e.g., mutations, disclosed in Table 21. In some embodiments, an anti-TCRVβ antibody disclosed herein comprise an Asn297Ala (N297A) mutation. In some embodiments, an anti-TCRVβ antibody disclosed herein comprise a Leu234A1a/Leu235Ala (LALA) mutation.

In some embodiments, an anti-TCRβV antibody molecule disclosed herein does not recognize, e.g., bind to, an interface of a TCRβV:TCRalpha complex.

In some embodiments, an anti-TCRβV antibody molecule disclosed herein does not recognize, e.g., bind to, a constant region of a TCRβV protein. An exemplary antibody that binds to a constant region of a TCRBV region is JOVI. 1 as described in Viney et al., (Hybridoma. 1992 December; 11(6):701-13).

In some embodiments, an anti-TCRβV antibody molecule disclosed herein does not recognize, e.g., bind to, one or more (e.g., all) of a complementarity determining region (e.g., CDR1, CDR2 and/or CDR3) of a TCRβV protein.

In some embodiments, binding of the anti-TCRβV antibody molecule to a TCRβV region results in one, two, three, four, five, six, seven, eight, nine, ten or more (e.g., all) of the following:

    • (i) reduced level, e.g., expression level, and/or activity of IL-1(3;
    • (ii) reduced level, e.g., expression level, and/or activity of IL-6;
    • (iii) reduced level, e.g., expression level, and/or activity of TNFα;
    • (iv) increased level, e.g., expression level, and/or activity of IL-2;
    • (v) a delay, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more hours delay, in increased level, e.g., expression level, and/or activity of IL-2;
    • (vi) a delay, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 hours delay, in increased level, e.g., expression level, and/or activity of IFN-gamma;
    • (vii) reduced T cell proliferation kinetics;
    • (viii) reduced cytokine storm, e.g., cytokine release syndrome (CRS) and/or neurotoxicity (NT), e.g., as measured by an assay of Example 3;
    • (ix) cell killing, e.g., target cell killing, e.g. cancer cell killing, e.g., as measured by an assay of Example 4;
    • (x) increased level, e.g., expression level, and/or activity of IL-15; or
    • (xi) increased Natural Killer (NK) cell proliferation, e.g., expansion.

In some embodiments, any one or all of (i)-(xi) or any combination thereof resulting from an anti-TCRβV antibody molecule disclosed herein is compared to an antibody that binds to: a CD3 molecule, e.g., CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments, binding of the anti-TCRβV antibody molecule to a TCRβV region results in secretion, e.g., production of perforin and/or Granzyme B.

In an aspect, the disclosure provides an antibody molecule which binds, e.g., specifically binds, to a T cell receptor beta variable chain (TCRβV) region, wherein the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (a) a light chain variable region (VL) comprising:
    • (i) one, two or all of (e.g., three) a light chain complementarity determining region 1 (LC CDR1), a light chain complementarity determining region 2 (LC CDR2), and a light chain complementarity determining region 3 (LC CDR3) of SEQ ID NO: 10 or SEQ ID NO: 11; and
    • (ii) a framework region (FR) having at least 95% identity with one, two, three, or all of (e.g., four) a non-murine germline framework 1 (FR1), a non-murine germline framework region 2 (FR2), a non-murine germline framework region 3 (FR3), and a non-murine germline framework region 4 (FR4); and/or
    • (b) a heavy chain variable region (VH) comprising:
    • (i) one, two or all of (e.g., three) a heavy chain complementarity determining region 1 (HC CDR1), a heavy chain complementarity determining region 2 (HC CDR2) and a heavy chain complementarity determining region 3 (HC CDR3) of SEQ ID NO: 9; and
    • (ii) a framework region (FR) having at least 95% identity with one, two, three, or all of (e.g., four) a non-murine germline framework 1 (FR1), a non-murine germline framework region 2 (FR2), a non-murine germline framework region 3 (FR3), and a non-murine germline framework region 4 (FR4).

In some embodiments, the VL comprises a sequence having a consensus sequence of SEQ ID NO: 230 or 3289.

In some embodiments, the VH comprises a sequence having a consensus sequence of SEQ ID NO: 231 or 3290.

In some embodiments, the anti-TCRβV antibody molecule binds to TCRβ V6, e.g., one or more of TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01, or a variant thereof.

In some embodiment, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of SEQ ID NO: 1 or SEQ ID NO: 9, or an amino acid sequence listed in Table 1; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11, or an amino acid sequence listed in Table 1.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11, or an amino acid sequence listed in Table 1.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of SEQ ID NO:1 or SEQ ID NO: 9, or an amino acid sequence listed in Table 1.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a VL comprising: a LC CDR1 amino acid sequence of SEQ ID NO: 6 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), a LC CDR2 amino acid sequence of SEQ ID NO:7 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), and/or a LC CDR3 amino acid sequence of SEQ ID NO:8 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof); and/or
    • (ii) a VH comprising: a HC CDR1 amino acid sequence of SEQ ID NO: 3 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), a HC CDR2 amino acid sequence of SEQ ID NO:4 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), and/or a HC CDR3 amino acid sequence of SEQ ID NO:5 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof).

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • a variable heavy chain (VH) of an amino acid sequence listed in Table 1, e.g., SEQ ID NO: 9, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to an amino acid sequence listed in Table 1, e.g., SEQ ID NO: 9 or SEQ ID NO: 1312; and/or
    • a variable light chain (VL) of an amino acid sequence listed in Table 1, e.g., SEQ ID NO: 10 or SEQ ID NO: 11, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to an amino acid sequence listed in Table 1, e.g., SEQ ID NO: 10 or SEQ ID NO: 11 or SEQ ID NO: 1314.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) the VH amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 1312;
    • (ii) an amino acid sequence having at least about 85%, 90%, 95%, or 99% sequence identity to the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 1312;
    • (iii) the VL amino acid sequence of SEQ ID NO: 10 or SEQ ID NO: 1314; and/or
    • (iv) an amino acid sequence having at least about 85%, 90%, 95%, or 99% sequence identity to the amino acid sequence of SEQ ID NO: 10 or SEQ ID NO: 1314.

In an aspect, provided herein is an antibody molecule which binds, e.g., specifically binds, to a T cell receptor beta variable chain (TCRβV) region, wherein the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (a) a light chain variable region (VL) comprising:
    • (i) one, two or all of (e.g., three) a light chain complementarity determining region 1 (LC CDR1), a light chain complementarity determining region 2 (LC CDR2), and a light chain complementarity determining region 3 (LC CDR3) of a humanized B-H light chain (LC) of Table 2; and
    • (ii) a framework region (FR) having at least 95% identity with one, two, three or all (e.g., four) of a framework region 1 (FR1), a framework region 2 (FR2), a framework region 3 (FR3), and a framework region 4 (FR4) of a humanized B-H LC of Table 2; and/or
    • (b) a heavy chain variable region (VH) comprising:
    • (i) one, two or all of (e.g., three) a heavy chain complementarity determining region 1 (HC CDR1), a heavy chain complementarity determining region 2 (HC CDR2) and a heavy chain complementarity determining region 3 (HC CDR3) of a humanized B-H heavy chain (HC) of Table 2; and
    • (ii) a framework region (FR) having at least 95% identity with one, two, three or all (e.g., four) of a framework region 1 (FR1), a framework region 2 (FR2), a framework region 3 (FR3), and a framework region 4 (FR4) of a humanized B-H HC of Table 2.

In some embodiments, the anti-TCRβV binds to TCRβ V12, e.g., TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01, or a variant thereof.

In some embodiment, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of Antibody B listed in Table 2; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of Antibody B listed in Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of a humanized B-H antibody listed in Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of a humanized B-H antibody listed in Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of a humanized B-H antibody listed in Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • a VH sequence of a humanized B-H antibody listed in Table 2, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VH of a humanized B-H antibody listed in Table 2; and/or
    • a VL sequence of a humanized B-H antibody listed in Table 2, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VL of a humanized B-H antibody listed in Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with one of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H LC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with any two of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H LC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with any three of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H LC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with all of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H LC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with one of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H HC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with any two of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H HC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with any three of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H HC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with all of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H HC of Table 2.

In some embodiment, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of Antibody C listed in Table 10; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of Antibody C listed in Table 10.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of antibody C or humanized C-H antibody listed in Table 10.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of antibody C or humanized C-H antibody listed in Table 10.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • a VH sequence of a humanized C-H antibody listed in Table 10, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VH of a humanized C-H antibody listed in Table 10; and/or
    • a VL sequence of a humanized C-H antibody listed in Table 10, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VL of a humanized C-H antibody listed in Table 10.

In some embodiment, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of Antibody E listed in Table 11; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of Antibody E listed in Table 11.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of antibody E or humanized E-H antibody listed in Table 11.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of antibody E or humanized E-H antibody listed in Table 11.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • a VH sequence of a humanized E-H antibody listed in Table 11, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VH of a humanized E-H antibody listed in Table 11; and/or
    • a VL sequence of a humanized E-H antibody listed in Table 11, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VL of a humanized E-H antibody listed in Table 11.

In some embodiment, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of Antibody D listed in Table 12; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of Antibody D listed in Table 12.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of antibody D or humanized D-H antibody listed in Table 12.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of antibody D or humanized D-H antibody listed in Table 12.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • a VH sequence of a humanized D-H antibody listed in Table 12, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VH of a humanized D-H antibody listed in Table 12; and/or
    • a VL sequence of a humanized D-H antibody listed in Table 12, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VL of a humanized D-H antibody listed in Table 12.

In some embodiment, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of Antibody G listed in Table 13; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of Antibody G listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of antibody G or humanized G-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of antibody G or humanized G-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • a VH sequence of a humanized G-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VH of a humanized G-H antibody listed in Table 13; and/or
    • a VL sequence of a humanized G-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VL of a humanized G-H antibody listed in Table 13.

In some embodiment, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of Antibody H listed in Table 13; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of Antibody H listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of antibody H or humanized H-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of antibody H or humanized H-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • a VH sequence of a humanized H-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VH of a humanized H-H antibody listed in Table 13; and/or
    • a VL sequence of a humanized H-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VL of a humanized H-H antibody listed in Table 13.

In some embodiment, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of Antibody I listed in Table 13; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of Antibody I listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of antibody I or humanized I-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of antibody I or humanized I-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • a VH sequence of a humanized I-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VH of a humanized I-H antibody listed in Table 13; and/or
    • a VL sequence of a humanized I-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VL of a humanized I-H antibody listed in Table 13.

In some embodiment, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of Antibody J listed in Table 13; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of Antibody J listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of antibody J or humanized J-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of antibody J or humanized J-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • a VH sequence of a humanized J-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VH of a humanized J-H antibody listed in Table 13; and/or
    • a VL sequence of a humanized J-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VL of a humanized J-H antibody listed in Table 13.

In some embodiment, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of Antibody K listed in Table 13; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of Antibody K listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of antibody K or humanized K-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of antibody K or humanized K-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • a VH sequence of a humanized G-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VH of a humanized K-H antibody listed in Table 13; and/or
    • a VL sequence of a humanized G-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VL of a humanized K-H antibody listed in Table 13.

In some embodiment, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of Antibody L listed in Table 13; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of Antibody L listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of antibody L or humanized L-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of antibody L or humanized L-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • a VH sequence of a humanized L-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VH of a humanized L-H antibody listed in Table 13; and/or
    • a VL sequence of a humanized L-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VL of a humanized L-H antibody listed in Table 13.

In some embodiment, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of Antibody M listed in Table 13; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of Antibody M listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of antibody M or humanized M-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of antibody M or humanized M-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • a VH sequence of a humanized M-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VH of a humanized M-H antibody listed in Table 13; and/or
    • a VL sequence of a humanized M-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VL of a humanized M-H antibody listed in Table 13.

In some embodiment, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of Antibody N listed in Table 13; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of Antibody N listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of antibody N or humanized N-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of antibody N or humanized N-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • a VH sequence of a humanized N-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VH of a humanized N-H antibody listed in Table 13; and/or
    • a VL sequence of a humanized N-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VL of a humanized N-H antibody listed in Table 13.

In some embodiment, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of Antibody 0 listed in Table 13; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of Antibody 0 listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of antibody 0 or humanized O-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of antibody 0 or humanized O-H antibody listed in Table 13.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • a VH sequence of a humanized O-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VH of a humanized O-H antibody listed in Table 13; and/or
    • a VL sequence of a humanized O-H antibody listed in Table 13, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VL of a humanized O-H antibody listed in Table 13.

In another aspect, the disclosure provides a non-murine, e.g., a human-like antibody molecule (e.g., a human or humanized antibody molecule), which binds, e.g., specifically binds, to a T cell receptor beta variable (TCRβV) region. In some embodiments, binding of the anti-TCRβV antibody molecule results in expansion, e.g., at least about 1.1-50 fold expansion (e.g., at least about 1.5-40 fold, 2-35 fold, 3-30 fold, 5-25 fold, 8-20 fold, or 10-15 fold expansion), of a population of T cells, e.g., a population of T cells having a memory-like phenotype, e.g., CD45RA+ CCR7—T cells. In some embodiments, the population of T cells having a memory-like phenotype comprises CD4+ and/or CD8+ T cells. In some embodiments, the population of T cells having a memory-like phenotype comprises a population of memory T cells, e.g., T effector memory (TEM) cells, e.g., TEM cells expressing CD45RA (TEMRA) cells, e.g., CD4+ or CD8+ TEMRA cells. In some embodiments, the population of T cells having a memory-like phenotype does not express a senescent marker, e.g., CD57. In some embodiments, the population of T cells having a memory-like phenotype does not express an inhibitory receptor, e.g., OX40, 4-1BB, and/or ICOS.

In some embodiments, the population of T cells having a memory-like phenotype is a population of T cells with CD45RA+ CCR7− CD57−. In some embodiments, the population of T cells having a memory-like phenotype does not express an inhibitory receptor, e.g., OX40, 4-1BB, and/or ICOS.

In some embodiments, the population of T cells having a memory-like phenotype, e.g., as described herein, has increased proliferative capacity, e.g., as compared to a reference cell population, e.g., an otherwise similar population of cells that has not been contacted with an anti-TCRβV antibody.

In some embodiments, the expansion is at least about 1.1-10 fold expansion (e.g., at least about 1.1, 1.2, 1.3, 1.4, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, or 10 fold expansion).

In some embodiments, expansion of the population of T cells having a memory-like phenotype, e.g., memory effector T cells, e.g., TEM cells, e.g., TEMRA cells, e.g., CD4+ or CD8+ TEMRA cells, is compared to expansion of a similar population of cells with an antibody that binds to: a CD3 molecule, e.g., CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments, the population of expanded T cells having a memory-like phenotype, e.g., T effector memory cells, comprises cells T cells, e.g., CD3+, CD8+ or CD4+ T cells. In some embodiments, the population of expanded T cells having a memory-like phenotype, T effector memory cells, comprises CD3+ and CD8+ T cells. In some embodiments, the population of expanded T cells having a memory-like phenotype, e.g., T effector memory cells comprises CD3+ and CD4+ T cells.

In some embodiments, the population of expanded T cells having a memory-like phenotype, T effector memory (TEM) cells, comprises cells T cells, e.g., CD3+, CD8+ or CD4+ T cells, which express or re-express, CD45RA, e.g., CD45RA+. In some embodiments, the population comprises TEM cells expressing CD45RA, e.g., TEMRA cells. In some embodiments, expression of CD45RA on TEMRA cells, e.g., CD4+ or CD8+ TEMRA cells, can be detected by a method disclosed herein, e.g., flow cytometry.

In some embodiments, the population of T cells having a memory-like phenotype, e.g., TEMRA cells have low or no expression of CCR7, e.g., CCR7− or CCR7 low. In some embodiments, expression of CCR7 on TEMRA cells cannot be detected by a method disclosed herein, e.g., flow cytometry.

In some embodiments, the population of T cells having a memory-like phenotype, e.g., TEMRA cells express CD95, e.g., CD95+. In some embodiments, expression of CD95 on TEMRA cells can be detected by a method disclosed herein, e.g., flow cytometry.

In some embodiments, the population of T cells having a memory-like phenotype, e.g., TEMRA cells express CD45RA, e.g., CD45RA+, have low or no expression of CCR7, e.g., CCR7− or CCR7 low, and express CD95, e.g., CD95+. In some embodiments, the population of T cells having a memory-like phenotype, e.g., TEMRA cells can be identified as CD45RA+, CCR7− and CD95+ cells. In some embodiments, the population of T cells having a memory-like phenotype, e.g., TEMRA cells comprise CD3+, CD4+ or CD8+ T cells (e.g., CD3+ T cells, CD3+ CD8+ T cells, or CD3+CD4+ T cells).

In some embodiments, the population of T cells having a memory-like phenotype does not express a senescent marker, e.g., CD57.

In some embodiments, the population of T cells having a memory-like phenotype does not express an inhibitory receptor, e.g., OX40, 4-1BB, and/or ICOS.

In some embodiments, binding of the anti-TCRβV antibody molecule results in expansion, e.g., at least about 1.1-50 fold expansion (e.g., at least about 1.5-40 fold, 2-35 fold, 3-30 fold, 5-25 fold, 8-20 fold, or 10-15 fold expansion), of a subpopulation of T cells. In some embodiments, the anti-TCRβV antibody molecule-activated (e.g., expanded) subpopulation of T cells resemble TEMRA cells in high expression of CD45RA and/or low expression of CCR7. In some embodiments, the anti-TCRβV antibody molecule-activated (e.g., expanded) subpopulation of T cells do not display upregulation of the senescence markers CD57 and/or KLRG1. In some embodiments, the anti-TCRβV antibody molecule-activated (e.g., expanded) subpopulation of T cells do not display upregulation of co-stimulatory molecules CD27 and/or CD28. In some embodiments, the anti-TCRβV antibody molecule-activated (e.g., expanded) subpopulation of T cells are highly proliferative. In some embodiments, the anti-TCRβV antibody molecule-activated (e.g., expanded) subpopulation of T cells secrete IL-2. In some embodiments, expression of surface markers on T cells can be detected by a method disclosed herein, e.g., flow cytometry. In some embodiments, the proliferative capability of T cells can be detected by a method disclosed herein, e.g., a method described in Example 4. In some embodiments, cytokine expression of T cells can be detected by a method disclosed herein, e.g., a method described in Examples 10 and 21. In some embodiments, the expansion is at least about 1.1-10 fold expansion (e.g., at least about 1.1, 1.2, 1.3, 1.4, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, or 10 fold expansion). In some embodiments, the expansion is compared to expansion of a similar population of cells with an antibody that binds to a CD3 molecule, e.g., CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments, binding of the anti-TCRβV antibody molecule to a TCRβV region results in one, two, three, four, five, six, seven, eight, nine, ten or more (e.g., all) of the following:

    • (i) reduced level, e.g., expression level, and/or activity of IL-1β;
    • (ii) reduced level, e.g., expression level, and/or activity of IL-6;
    • (iii) reduced level, e.g., expression level, and/or activity of TNFα;
    • (iv) increased level, e.g., expression level, and/or activity of IL-2;
    • (v) a delay, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more hours delay, in increased level, e.g., expression level, and/or activity of IL-2;
    • (vi) a delay, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 hours delay, in increased level, e.g., expression level, and/or activity of IFNg;
    • (vii) reduced T cell proliferation kinetics;
    • (viii) reduced cytokine storm, e.g., cytokine release syndrome (CRS) and/or neurotoxicity (NT), e.g., as measured by an assay of Example 3;
    • (ix) cell killing, e.g., target cell killing, e.g. cancer cell killing, e.g., as measured by an assay of Example 4;
    • (x) increased level, e.g., expression level, and/or activity of IL-15; or
    • (xi) increased Natural Killer (NK) cell proliferation, e.g., expansion, compared to an antibody that binds to: a CD3 molecule, e.g., CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments of any of the compositions disclosed herein, binding of the anti-TCRβV antibody molecule to a TCRβV region results in a reduction of at least 2, 5, 10, 20, 50, 100, or 200 fold, or at least 2-200 fold (e.g., 5-150, 10-100, 20-50 fold) in the expression level and or activity of IL-10 as measured by an assay of Example 3.

In some embodiments of any of the compositions disclosed herein, binding of the anti-TCRβV antibody molecule to a TCRβV region results in a reduction of at least 2, 5, 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 fold, or at least 2-1000 fold (e.g., 5-900, 10-800, 20-700, 50-600, 100-500, or 200-400 fold) in the expression level and or activity of IL-6 as measured by an assay of Example 3.

In some embodiments of any of the compositions disclosed herein, binding of the anti-TCRβV antibody molecule to a TCRβV region results in a reduction of at least 2, 5, 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, or 2000 fold, or at least 2-2000 fold (e.g., 5-1000, 10-900, 20-800, 50-700, 100-600, 200-500, or 300-400 fold) in the expression level and or activity of TNFα as measured by an assay of Example 3.

In some embodiments of any of the compositions disclosed herein, binding of the anti-TCRβV antibody molecule to a TCRβV region results in an increase of at least 2, 5, 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, or 2000 fold, or at least 2-2000 fold (e.g., 5-1000, 10-900, 20-800, 50-700, 100-600, 200-500, or 300-400 fold) in the expression level and or activity of IL-2 as measured by an assay of Example 3.

In some embodiments of any of the compositions disclosed herein, binding of the anti-TCRβV antibody molecule to a TCRβV region results in an increase of at least 2, 5, 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, or 2000 fold, or at least 2-2000 fold (e.g., 5-1000, 10-900, 20-800, 50-700, 100-600, 200-500, or 300-400 fold) in the expression level and or activity of IL-15 as measured by an assay of Example 4.

In some embodiments of any of the compositions disclosed herein, binding of the anti-TCRβV antibody molecule results in proliferation, e.g., expansion, e.g., at least about 1.1-50 fold expansion (e.g., at least about 1.5-40 fold, 2-35 fold, 3-30 fold, 5-25 fold, 8-20 fold, or 10-15 fold expansion), of a population of Natural Killer (NK) cells. In some embodiments, the expansion of NK cells is at least about 1.1-30 fold expansion (e.g., at least about 1.1, 1.2, 1.3, 1.4, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, or at least about 1.1-5, 5-10, 10-15, 15-20, 20-25, or 25-30 fold expansion). In some embodiments, the expansion of NK cells is measure by an assay of Example 4. In some embodiments, the expansion of NK cells by, e.g., binding of, the anti-TCRβV antibody molecule is compared to expansion of an otherwise similar population not contacted with the anti-TCRβV antibody molecule.

In some embodiments of any of the compositions disclosed herein, binding of the anti-TCRβV antibody molecule results in cell killing, e.g., target cell killing, e.g. cancer cell killing. In some embodiments, the cancer cell is a hematological cancer cell or a solid tumor cell. In some embodiments, the cancer cell is a multiple myeloma cell. In some embodiments, binding of the anti-TCRβV antibody molecule results in cell killing in vitro or in vivo. In some embodiments, cell killing is measured by an assay of Example 4.

In some embodiments of any of the compositions disclosed herein, binding of the anti-TCRβV antibody molecule to a TCRβV region results in an increase or decrease of at least 2, 5, 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, or 2000 fold, or at least 2-2000 fold (e.g., 5-1000, 10-900, 20-800, 50-700, 100-600, 200-500, or 300-400 fold) of any of the activities described herein compared the activity of 16G8 or TM23 murine antibody, or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In an aspect, provided herein is an antibody molecule which binds, e.g., specifically binds, to a T cell receptor beta variable chain (TCRβV) region (an anti-TCRβV antibody molecule), wherein the anti-TCRβV antibody molecule:

    • (i) binds specifically to an epitope on TCRβV, e.g., the same or similar epitope as the epitope recognized by an anti-TCRβV antibody molecule as described herein, e.g., a second anti-TCRβV antibody molecule;
    • (ii) shows the same or similar binding affinity or specificity, or both, as an anti-TCRβV antibody molecule as described herein, e.g., a second anti-TCRβV antibody molecule;
    • (iii) inhibits, e.g., competitively inhibits, the binding of an anti-TCRβV antibody molecule as described herein, e.g., a second anti-TCRβV antibody molecule;
    • (iv) binds the same or an overlapping epitope with an anti-TCRβV antibody molecule as described herein, e.g., a second anti-TCRβV antibody molecule; or
    • (v) competes for binding, and/or binds the same epitope, with an anti-TCRβV antibody molecule as described herein, e.g., a second anti-TCRβV antibody molecule,
    • In some embodiments, the second anti-TCRβV antibody molecule comprises an antigen binding domain chosen from Table 1 or Table 2, or a sequence substantially identical thereto. In some embodiments, the second anti-TCRβV antibody molecule comprises an antigen binding domain, comprising:
    • a heavy chain complementarity determining region 1 (HC CDR1), a heavy chain complementarity determining region 2 (HC CDR2) and/or a heavy chain complementarity determining region 3 (HC CDR3) of SEQ ID NO: 1 or SEQ ID NO: 9; and/or a light chain complementarity determining region 1 (LC CDR1), a light chain complementarity determining region 2 (LC CDR2), and/or a light chain complementarity determining region 3 (LC CDR3) of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11.

In some embodiments of any of the compositions disclosed herein, binding of the anti-TCRβV antibody molecule to a TCRβV region results in a change in any (e.g., one, two, three, four or all) of (i)-(v) that is different, e.g., an increase or decrease, of at least 2, 5, 10, 20, 50, 100-fold, compared the activity of 16G8 or TM23 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments of any of the compositions disclosed herein, the anti-TCRβV antibody molecule binds to a TCRBV family (e.g., gene family), e.g., a TCRBV gene family comprising subfamilies, e.g., as described herein. In some embodiments, the TCRBV family, e.g., gene family, comprises: a TCRβ V6 subfamily, a TCRβ V10 subfamily, a TCRβ V12 subfamily, a TCRβ V5 subfamily, a TCRβ V7 subfamily, a TCRβ V11 subfamily, a TCRβ V14 subfamily, a TCRβ V16 subfamily, a TCRβ V18 subfamily, a TCRβ V9 subfamily, a TCRβ V13 subfamily, a TCRβ V4 subfamily, a TCRβ V3 subfamily, a TCRβ V2 subfamily, a TCRβ V15 v, a TCRβ V30 subfamily, a TCRβ V19 subfamily, a TCRβ V27 subfamily, a TCRβ V28 subfamily, a TCRβ V24 subfamily, a TCRβ V20 subfamily, TCRβ V25 subfamily, a TCRβ V29 subfamily, a TCRβ V23 subfamily, a TCRβ V21 subfamily, a TCRβ V1 subfamily, a TCRβ V17 subfamily, or a TCRβ V26 subfamily.

In some embodiments, the anti-TCRβV antibody binds to a TCRβ V6 subfamily chosen from: TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01. In some embodiments the TCRβ V6 subfamily comprises TCRβ V6-5*01.

In some embodiments, the anti-TCRβV antibody binds to a TCRβ V10 subfamily chosen from: TCRβ V10-1*01, TCRβ V10-1*02, TCRβ V10-3*01 or TCRβ V10-2*01.

In some embodiments, the anti-TCRβV antibody binds to a TCRβ V12 subfamily chosen from: TCRβ V12-4*01, TCRβ V12-3*01 or TCRβ V12-5*01.

In some embodiments of any of the compositions disclosed herein, the anti-TCRβV antibody molecule does not bind to TCRβ V12, or binds to TCRβ V12 with an affinity and/or binding specificity that is less than (e.g., less than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the 16G8 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments of any of the compositions disclosed herein, the anti-TCRβV antibody molecule binds to TCRβ V12 with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the 16G8 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments of any of the compositions disclosed herein, the anti-TCRβV antibody molecule binds to a TCRβV region other than TCRβ V12 (e.g., TCRβV region as described herein, e.g., TCRβ V6 subfamily (e.g., TCRβ V6-5*01) with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the 16G8 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments of any of the compositions disclosed herein, the anti-TCRβV antibody molecule does not comprise at least one CDR of Antibody B. In some embodiments of any of the compositions disclosed herein, the anti-TCRβV antibody molecule does not comprise the CDRs of Antibody B.

In some embodiments of any of the compositions disclosed herein, the anti-TCRβV antibody binds to a TCRβ V5 subfamily chosen from: TCRβ V5-5*01, TCRβ V5-6*01, TCRβ V5-4*01, TCRβ V5-8*01, TCRβ V5-1*01.

In some embodiments of any of the compositions disclosed herein, the anti-TCRβV antibody binds to a TCRβ V5 subfamily chosen from: TCRβ V5-5*01, TCRβ V5-6*01, TCRβ V5-4*01, TCRβ V5-8*01, TCRβ V5-1*01.

In some embodiments of any of the compositions disclosed herein, the anti-TCRβV antibody molecule does not bind to TCRβ V5-5*01 or TCRβ V5-1*01, or binds to TCRβ V5-5*01 or TCRβ V5-1*01 with an affinity and/or binding specificity that is less than (e.g., less than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the TM23 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments of any of the compositions disclosed herein, the anti-TCRβV antibody molecule binds to TCRβ V5-5*01 or TCRβ V5-1*01 with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the TM23 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments of any of the compositions disclosed herein, the anti-TCRβV antibody molecule binds to a TCRβV region other than TCRβ V5-5*01 or TCRβ V5-1*01 (e.g., TCRβV region as described herein, e.g., TCRβ V6 subfamily (e.g., TCRβ V6-5*01) with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the TM23 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments of any of the compositions disclosed herein, the anti-TCRβV antibody molecule does not comprise at least one CDR of the TM23 murine antibody. In some embodiments of any of the compositions disclosed herein, the anti-TCRβV antibody molecule does not comprise the CDRs of the TM23 murine antibody.

In some embodiments of any of the compositions disclosed herein, an anti-TCRβV antibody molecule disclosed herein does not comprise the sequence of a murine anti-rat TCR antibody R73, e.g., as disclosed in J Exp Med. 1989 Jan. 1; 169(1): 73-86, herein incorporated by reference in its entirety. In some embodiments of any of the compositions disclosed herein, a multispecific antibody molecule disclosed herein does not comprise the sequence of a murine anti-rat TCR antibody R73, e.g., as disclosed in J Immunol. 1993 Mar. 15; 150(6):2305-15, herein incorporated by reference in its entirety.

In some embodiments of any of the compositions disclosed herein, an anti-TCRβV antibody molecule disclosed herein does not comprise a viral peptide-MHC complex, e.g., as disclosed in Oncoimmunology. 2016; 5(1): e1052930, herein incorporated by reference in its entirety. In some embodiments of any of the compositions disclosed herein, a multispecific antibody molecule disclosed herein does not comprise a viral peptide-MHC complex, e.g., as disclosed in Oncoimmunology. 2016; 5(1): e1052930, herein incorporated by reference in its entirety.

In some embodiments of any of the compositions disclosed herein, the anti-TCRβV antibody molecule binds to one or more (e.g., all) of the following TCRβV subfamilies:

    • (i) TCRβ V6 subfamily comprising, e.g., one or more of TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01;
    • (ii) TCRβ V10 subfamily comprising, e.g., one or more of TCRβ V10-1*01, TCRβ V10-1*02, TCRβ V10-3*01 or TCRβ V10-2*01;
    • (iii) TCRβ V5 subfamily comprising, e.g., one or more of TCRβ V5-6*01, TCRβ V5-4*01, or TCRβ V5-8*01;
    • (iv) TCRβ V12 subfamily comprising e.g., one or more of TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01;
    • (v) TCRβ V7 subfamily comprising e.g., one or more of TCRβ V7-7*01, TCRβ V7-6*01, TCRβ V7-8*02, TCRβ V7-4*01, TCRβ V7-2*02, TCRβ V7-2*03, TCRβ V7-2*01, TCRβ V7-3*01, TCRβ V7-9*03, or TCRβ V7-9*01;
    • (vi) TCRβ V11 subfamily comprising e.g., one or more of TCRβ V11-1*01, TCRβ V11-2*01 or TCRβ V11-3*01;
    • (vii) TCRβ V14 subfamily comprising TCRβ V14*01;
    • (viii) TCRβ V16 subfamily comprising TCRβ V16*01;
    • (ix) TCRβ V18 subfamily comprising TCRβ V18*01;
    • (x) TCRβ V9 subfamily comprising T e.g., one or more of CRP V9*01 or TCRβ V9*02;
    • (xi) TCRβ V13 subfamily comprising TCRβ V13*01;
    • (xii) TCRβ V4 subfamily comprising e.g., one or more of e.g., one or more of TCRβ V4-2*01, TCRβ V4-3*01, or TCRβ V4-1*01;
    • (xiii) TCRβ V3 subfamily comprising TCRβ V3-1*01;
    • (xiv) TCRβ V2 subfamily comprising TCRβ V2*01;
    • (xv) TCRβ V15 subfamily comprising TCRβ V15*01;
    • (xvi) TCRβ V30 subfamily comprising e.g., one or more of TCRβ V30*01, or TCRβ V30*02;
    • (xvii) TCRβ V19 subfamily comprising e.g., one or more of TCRβ V19*01, or TCRβ V19*02;
    • (xviii) TCRβ V27 subfamily comprising TCRβ V27*01;
    • (xix) TCRβ V28 subfamily comprising TCRβ V28*01;
    • (xx) TCRβ V24 subfamily comprising TCRβ V24-1*01;
    • (xxi) TCRβ V20 subfamily comprising e.g., one or more of TCRβ V20-1*01, or TCRβ V20-1*02;
    • (xxii) TCRβ V25 subfamily comprising TCRβ V25-1*01; or
    • (xxiii) TCRβ V29 subfamily comprising TCRβ V29-1*01;
    • (xxiv) TCRβ V21 subfamily;
    • (xxv) TCRβ V1 subfamily;
    • (xxvi) TCRβ V17 subfamily;
    • (xvii) TCRβ V23 subfamily; or
    • (xviii) TCRβ V26 subfamily.

In some embodiments of any of the compositions disclosed herein, the anti-TCRβV antibody molecule binds to one or more (e.g., all) of the following TCRβV subfamilies:

    • (i) TCRβ V6, e.g., one or more of TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01;
    • (ii) TCRβ V10, e.g., one or more of TCRβ V10-1*01, TCRβ V10-1*02, TCRβ V10-3*01 or TCRβ V10-2*01;
    • (iii) TCRβ V12, e.g., one or more of TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01; or
    • (iv) TCRβ V5, e.g., one or more of TCRβ V5-5*01, TCRβ V5-6*01, TCRβ V5-4*01, TCRβ V5-8*01, TCRβ V5-1*01.

In some embodiments, the anti-TCRβV antibody molecule binds to TCRβ V6, e.g., one or more of TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01. In some embodiments, the anti-TCRβV antibody molecule binds to TCRβ V6-5*01.

In some embodiments, the anti-TCRβV antibody molecule does not bind to TCRβ V12.

In some embodiments, the anti-TCRβV antibody molecule does not bind to TCRβ V5-5*01 or TCRβ V5-1*01.

In an aspect, provided herein is a multispecific molecule (e.g., a bispecific molecule), comprising a first moiety (e.g., a first immune cell engager) comprising an antibody molecule which binds (e.g., specifically binds) to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”).

In some embodiments, the multispecific molecule comprises a second moiety which comprises one or more of: a tumor-targeting moiety, a cytokine molecule, a stromal modifying moiety, or an anti-TCRβV antibody molecule other than the first moiety.

In some embodiments, binding of the first moiety to the TCRβV region results in a cytokine profile, e.g., cytokine secretion profile, that differs from that of a T cell engager that binds to a receptor or molecule other than a TCRβV region (“a non-TCRβV-binding T cell engager”).

In another aspect, the disclosure provides a multispecific molecule, e.g., a bispecific molecule, comprising the anti-TCRβV antibody molecule disclosed herein.

In some embodiments, the multispecific molecule further comprises: a tumor-targeting moiety, a cytokine molecule, an immune cell engager, e.g., a second immune cell engager, and/or a stromal modifying moiety.

In yet another aspect, disclosed herein is a multispecific molecule, e.g., a bispecific molecule, comprising:

    • (i) a first moiety comprising a first immune cell engager comprising an anti-TCRβV antibody molecule disclosed herein; and
    • (ii) a second moiety comprising one or more of: a tumor-targeting moiety; a second immune cell engager; a cytokine molecule or a stromal modifying moiety.

In another aspect, the disclosure provides an isolated nucleic acid molecule comprising a nucleotide sequence encoding an anti-TCRβV antibody molecule disclosed herein, or a nucleotide sequence having at least 75%, 80%, 85%, 90%, 95%, or 99% identity thereto.

In another aspect, the disclosure provides an isolated nucleic acid molecule comprising a nucleotide sequence encoding a multispecific molecule disclosed herein, or a nucleotide sequence having at least 75%, 80%, 85%, 90%, 95%, or 99% identity thereto.

In yet another aspect, the disclosure provides a vector, e.g., an expression vector, comprising a nucleotide sequence encoding an anti-TCRβV antibody molecule disclosed herein, or a nucleotide sequence having at least 75%, 80%, 85%, 90%, 95%, or 99% identity thereto.

In another aspect, the disclosure provides a vector, e.g., an expression vector, comprising a nucleotide sequence encoding a multispecific molecule disclosed herein, or a nucleotide sequence having at least 75%, 80%, 85%, 90%, 95%, or 99% identity thereto.

In one aspect, the disclosure provides a cell, e.g., host cell, e.g., a population of cells, comprising a nucleic acid molecule encoding an anti-TCRβV antibody molecule disclosed herein, or a nucleotide sequence having at least 75%, 80%, 85%, 90%, 95%, or 99% identity thereto. In some embodiments, the cell or population of cells comprising a nucleic acid molecule encoding anti-TCRβV antibody molecule, comprises: (i) a heavy chain comprising: a variable region (VH), e.g., a VH listed in Tables 1-2 or 10-13, or a sequence having at least 75%, 80%, 85%, 90%, 95%, or 99% identity thereto; and one or more heavy chain constant regions, e.g., as described herein; and/or (ii) a light chain comprising: a variable region (VL) e.g., a VL listed in Tables 1-2 or 10-13, or a sequence having at least 75%, 80%, 85%, 90%, 95%, or 99% identity thereto; and a light chain constant region, e.g., as described herein, e.g., a kappa chain constant region comprising the sequence of SEQ ID NO: 39, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto. In some embodiments, the cell or population of cells further comprises an IgJ heavy chain constant region or a fragment thereof. In some embodiments, the IgJ heavy chain constant region comprises the sequence of SEQ ID NO: 76 or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto. In some embodiments, the IgJ is comprised in, e.g., expressed in, the same cell or population of cells comprising, e.g., expressing, the anti-TCRβV antibody molecule, e.g., the heavy chain and/or the light chain of the anti-TCRβV antibody molecule. In some embodiments, the IgJ is expressed in a different cell or population of cells than the cell or population of cells comprising, e.g., expressing, the anti-TCRβV antibody molecule, e.g., the heavy chain and/or the light chain of the anti-TCRβV antibody molecule.

In one aspect, the disclosure provides a cell, e.g., host cell, e.g., a population of cells, comprising a nucleic acid molecule encoding a multispecific molecule disclosed herein, or a nucleotide sequence having at least 75%, 80%, 85%, 90%, 95%, or 99% identity thereto.

In one aspect, disclosed herein is an anti-TCRβV antibody molecule for use in the manufacture of a medicament for treating a disease, e.g., cancer, in a subject.

In one aspect, disclosed herein is a multispecific molecule comprising an anti-TCRβV antibody molecule for use in the manufacture of a medicament for treating a disease, e.g., cancer, in a subject.

In another aspect, the disclosure provides a method of making, e.g., producing, an anti-TCRβV antibody molecule, a multispecific molecule described herein, comprising culturing a host cell described herein, under suitable conditions. In some embodiments of a method of making a multispecific molecule, the conditions comprise, e.g., conditions suitable for gene expression and/or homo- or heterodimerization.

In another aspect, the disclosure provides a pharmaceutical composition comprising an anti-TCRβV antibody molecule, or a multispecific molecule described herein, and a pharmaceutically acceptable carrier, excipient, or stabilizer.

In an aspect, the disclosure provides a method of modulating, e.g., enhancing, an immune response in a subject comprising administering to the subject an effective amount of an antibody molecule which binds (e.g., specifically binds) to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”).

In an aspect, the disclosure provides a method of modulating, e.g., enhancing, an immune response in a subject comprising administering to the subject an effective amount of a multispecific molecule disclosed herein.

In some embodiments, the method comprises expanding, e.g., increasing the number of, an immune cell population in the subject.

In an aspect, the disclosure provides a method of expanding, e.g., increasing the number of, an immune cell population comprising, contacting the immune cell population with an effective amount of an antibody molecule which binds (e.g., specifically binds) to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”).

In an aspect, the disclosure provides a method of expanding, e.g., increasing the number of, an immune cell population comprising, contacting the immune cell population with an effective amount of a multispecific molecule disclosed herein.

In some embodiments, the expansion occurs in vivo or ex vivo (e.g., in vitro).

In some embodiments, the immune cell population comprises a TCRβV expressing cell, e.g., a TCRβV+ cell.

In some embodiments, the TCRβV expressing cell is a T cell, e.g., a CD8+ T cell, a CD3+ T cell or a CD4+ T cell.

In some embodiments, the immune cell population comprises a T cell (e.g., a CD4 T cell, or a CD8 T cell). In some embodiments, the immune cell population comprises a T cell having a memory-like phenotype, e.g., CD45RA+ CCR7−. In some embodiments, the immune cell population comprises an effector T cell or a memory T cell (e.g., a memory effector T cell (e.g., TEM cell, e.g., TEMRA cell), or a tumor infiltrating lymphocyte (TIL)).

In some embodiments, the immune cell population comprises a T cell, a Natural Killer cell, a B cell, or a myeloid cell.

In some embodiments, the immune cell population is obtained from a healthy subject.

In an aspect, provided herein is a method of treating a disease e.g., cancer, in a subject comprising administering to the subject an effective amount, e.g., a therapeutically effective amount, of an anti-TCRβV antibody molecule or a multispecific molecule comprising an anti-TCRβV antibody molecule disclosed herein, thereby treating the disease.

In a related aspect, provided herein is a composition comprising an anti-TCRβV antibody molecule or a multispecific molecule comprising an anti-TCRβV antibody molecule disclosed herein, for use in the treatment of a disease, e.g., cancer, in a subject.

In some embodiments, the disease is a cancer, e.g., a solid tumor or a hematological cancer, or a metastatic lesion.

In some embodiments, the method further comprises administering a second agent, e.g., therapeutic agent, e.g., as described herein. In some embodiments, second agent comprises a therapeutic agent (e.g., a chemotherapeutic agent, a biologic agent, hormonal therapy), radiation, or surgery. In some embodiments, therapeutic agent is selected from: a chemotherapeutic agent, or a biologic agent.

In another aspect, provided herein is a method of targeting, e.g., directing or re-directing, a therapy, e.g., treatment, to a T cell, e.g., in a subject, e.g., having a disease, e.g., cancer, comprising administering an effective amount of: (i) an anti-TCRβV antibody disclosed herein; and (ii) the therapy, e.g., a tumor targeting therapy (e.g., an antibody that binds to a cancer antigen), e.g., as described herein, thereby targeting the T cell.

In some embodiments, (i) and (ii) are conjugated, e.g., linked.

In some embodiments, (i) and (ii) are administered simultaneously or concurrently.

In some embodiments, the method results in: reduced cytokine release syndrome (CRS) (e.g., lesser duration of CRS or no CRS), or a reduced severity of CRS (e.g., absence of severe CRS, e.g., CRS grade 4 or 5) compared to administration of (ii) alone. In some embodiments, CRS is assessed by an assay of Example 3. In some embodiments, the method results in: reduced neurotoxicity (NT) (e.g., lesser duration of NT or no NT), or a reduced severity of NT (e.g., absence of severe NT) compared to administration of (ii) alone.

In yet another aspect, the disclosure provides, a method of targeting a T cell, e.g., in a subject having a disease, e.g., cancer, with an anti-TCRβV antibody disclosed herein or a multispecific molecule comprising an anti-TCRβV antibody disclosed herein.

In another aspect, the disclosure provides a method of treating, e.g., preventing or reducing, cytokine release syndrome (CRS) and/or neurotoxicity (NT) in a subject, e.g., CRS and/or NT associated with a treatment, e.g., a previously administered treatment, comprising administering to the subject an effective amount of an anti-TCRβV antibody disclosed herein or a multispecific molecule comprising an anti-TCRβV antibody disclosed herein, wherein, the subject has a disease, e.g., a cancer, thereby treating, e.g., preventing or reducing, CRS and/or NT in the subject.

In a related aspect, the disclosure provides a composition comprising an anti-TCRβV antibody disclosed herein or a multispecific molecule comprising an anti-TCRβV antibody disclosed herein, for use in the treatment, e.g., prevention or reduction, of cytokine release syndrome (CRS) and/or neurotoxicity (NT) in a subject, e.g., CRS and/or NT associated with a treatment, e.g., a previously administered treatment, comprising administering to the subject an effective amount of the anti-TCRβV antibody, wherein the subject has a disease, e.g., a cancer.

In some embodiments of a method or composition for use disclosed herein, the anti-TCRβV antibody is administered concurrently with or after the administration of the treatment associated with CRS and/or NT.

In another aspect, provided herein is a method of expanding, e.g., increasing the number of, an immune cell population comprising, contacting the immune cell population with an antibody molecule, e.g., humanized antibody molecule, which binds, e.g., specifically binds, to a T cell receptor beta variable chain (TCRβV) region (e.g., anti-TCRβV antibody molecule described herein or a multispecific molecule comprising an anti-TCRβV antibody molecule described herein), thereby expanding the immune cell population.

In some embodiments, the expansion occurs in vivo or ex vivo (e.g., in vitro).

In an aspect, provided herein is a method of evaluating a subject having a cancer, comprising acquiring a value of the status of a TCRβV molecule for the subject, wherein said value comprises a measure of the presence of, e.g., level or activity of, a TCRβV molecule in a sample from the subject, wherein the value of the status of a TCRβV molecule is higher, e.g., increased, in a sample from the subject compared to a reference value, e.g., a value from a healthy subject, e.g., a subject that does not have cancer.

In another aspect, the disclosure provides a method of treating a subject having a cancer, the method comprising (i) acquiring a value of the status of a TCRβV molecule for the subject, wherein said value comprises a measure of the presence of, e.g., level or activity of, a TCRβV molecule in a sample from the subject, and (ii) responsive to said value, administering an effective amount of an anti-TCRβV antibody molecule described herein (e.g., a TCRβV agonist) to the subject, thereby treating the cancer.

In some embodiments, the value is higher, e.g., increased, in a sample from the subject compared to a reference value, e.g., a value from a healthy subject, e.g., a subject that does not have cancer.

In a related aspect, the disclosure provides a composition comprising an anti-TCRβV antibody molecule for use in the treatment of a subject having a cancer, comprising (i) acquiring a value of the status of a TCRβV molecule for the subject, wherein said value comprises a measure of the presence of, e.g., level or activity of, a TCRβV molecule in a sample from the subject, and (ii) responsive to said value, administering an effective amount of an anti-TCRβV antibody molecule described herein (e.g., a TCRβV agonist) to the subject.

In an aspect, provided herein is method of evaluating a subject for the presence of a cancer, the method comprising:

    • (i) acquiring a value of the status of one or more TCRβV molecules for the subject, e.g., in a biological sample from the subject, wherein said value comprises a measure of the presence of, e.g., level or activity of, a TCRβV molecule in a sample from the subject, and
    • (ii) determining whether the value for the one or more TCRβV molecules is higher, e.g., increased, in a sample from the subject compared to a reference value, e.g., a value from a healthy subject, e.g., a subject that does not have cancer,
    • wherein a value that is higher, e.g., increased, in the subject relative to the reference, e.g., healthy subject, is indicative of the presence of cancer in the subject.

In another aspect, the disclosure provides, a method of treating a subject having cancer, the method comprising:

    • (i) acquiring a value of the status of one or more TCRβV molecules for the subject, e.g., in a biological sample from the subject, wherein said value comprises a measure of the presence of, e.g., level or activity of, a TCRβV molecule in a sample from the subject;
    • (ii) determining whether the value for the one or more TCRβV molecules is higher, e.g., increased, in a sample from the subject compared to a reference value, e.g., a value from a healthy subject, e.g., a subject that does not have cancer, and
    • (iii) if a value that is higher, e.g., increased, in the subject relative to the reference value is determined, administering an effective amount of an anti-TCRβV antibody molecule, e.g., as described herein (e.g., a TCRβV agonist), to the subject, thereby treating the cancer.

In a related aspect, provided herein is a composition comprising anti-TCRβV antibody molecule for use in a method of treating a subject having a cancer, comprising

    • (i) acquiring a value of the status of one or more TCRβV molecules for the subject, e.g., in a biological sample from the subject, wherein said value comprises a measure of the presence of, e.g., level or activity of, a TCRβV molecule in a sample from the subject;
    • (ii) determining whether the value for the one or more TCRβV molecules is higher, e.g., increased, in a sample from the subject compared to a reference value, e.g., a value from a healthy subject, e.g., a subject that does not have cancer, and
    • (iii) if a value that is higher, e.g., increased, in the subject relative to the reference value is determined, administering an effective amount of an anti-TCRβV antibody molecule, e.g., as described herein (e.g., a TCRβV agonist), to the subject.

In some embodiments of any of the methods of treatment, or composition for use disclosed herein, the status is indicative of the subject having cancer, or a symptom thereof.

In some embodiments of any of the methods of treatment or composition for use disclosed herein, the status is indicative of responsiveness to a therapy, e.g., a therapy comprising an anti-TCRβV antibody molecule, e.g., as described herein.

In some embodiments of any of the methods of treatment or composition for use disclosed herein, the value of the status is determined, e.g., measured, by an assay described herein.

In yet another aspect, provided herein is a method of treating a subject having a cancer, comprising administering to the subject an effective amount of an anti-TCRβV antibody molecule described herein, wherein the subject has a higher, e.g., increased, level or activity of one or more TCRBV molecules, e.g., as described herein, compared to a reference level or activity of one or more TCRBV molecules, e.g., in a healthy subject, e.g., a subject not having a cancer

In an aspect, the disclosure provides, method of treating a subject having a cancer, comprising

    • (i) isolating a biological sample from the subject; e.g., a peripheral blood sample, biopsy sample, or bone marrow sample; and
    • (ii) acquiring a value of the status of one or more TCRβV molecules for the subject, e.g., in the biological sample from the subject, wherein said value comprises a measure of the presence of, e.g., level or activity of, a TCRβV molecule in a sample from the subject compared to a reference value, e.g., a sample from a health subject,
    • wherein a value that is higher, e.g., increased, in the subject relative to the reference, e.g., healthy subject, is indicative of the presence of cancer in the subject,
    • (iii) contacting the biological sample with an anti-TCRβV antibody molecule, e.g., in vitro; and
    • (iv) administering the biological sample or a portion thereof from step (iii) to the subject.

In another aspect, provided herein is method of expanding a population of immune effector cells from a subject having a cancer, the method comprising:

    • (i) isolating a biological sample comprising a population of immune effector cells from the subject; e.g., a peripheral blood sample, biopsy sample, or bone marrow sample;
    • (ii) acquiring a value of the status of one or more TCRβV molecules for the subject, e.g., in the biological sample from the subject, wherein said value comprises a measure of the presence of, e.g., level or activity of, a TCRβV molecule in a sample from the subject compared to a reference value, e.g., a sample from a health subject,
    • wherein a value that is higher, e.g., increased, in the subject relative to the reference, e.g., healthy subject, is indicative of the presence of cancer in the subject, and
    • (iii) contacting the biological sample comprising a population of immune effector cells with an anti-TCRβV antibody molecule.

In some embodiments, the method further comprises administering the population of immune effector cells contacted with the anti-TCRβV antibody molecule to the subject.

In some embodiments, a method of expansion, or method of treatment, or composition for use disclosed herein comprises measuring T cell function (e.g., cytotoxic activity, cytokine secretion, or degranulation) in the population of immune effector cells, e.g., compared to a reference population, e.g., an otherwise similar population not contacted with the anti-TCRβV antibody molecule or a population of immune effector cells obtained from a healthy subject (e.g., a subject that does not have a cancer).

In some embodiments of any of the methods or composition for use disclosed herein, the biological sample comprising the population of immune effector cells is contacted with an anti-TCRβV antibody molecule that binds to the one or more TCRβV molecules (e.g., the same TCRβV molecule) identified as being higher, e.g., increased, in the biological sample.

In some embodiments of any of the methods or composition for use disclosed herein, the biological sample comprising the population of immune effector cells is contacted with an anti-TCRβV antibody molecule that does not bind to the one or more TCRβV molecules (e.g., a different TCRβV molecule) identified as being higher, e.g., increased, in the biological sample.

In another aspect, provided herein is a method of identifying one or more TCRβV molecules associated with a cancer, the method comprising:

    • (i) acquiring a status for a plurality of TCRβV molecules in a biological sample from a first subject having the disease and in a biological sample from a second subject not having the disease; and
    • (ii) determining whether the level or activity of one or more of the TCRβV molecules is higher, e.g., increased, in the first subject relative to the second subject;
    • thereby identifying one or more TCRβV molecules associated with the cancer.

In some embodiments of any of the methods or composition for use disclosed herein, the one or more of the TCRβV molecules comprises one or more, (e.g., all) of the following TCRβV subfamilies:

    • (i) TCRβ V6 subfamily comprising, e.g., one or more of TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01;
    • (ii) TCRβ V10 subfamily comprising, e.g., one or more of TCRβ V10-1*01, TCRβ V10-1*02, TCRβ V10-3*01 or TCRβ V10-2*01;
    • (iii) TCRβ V5 subfamily comprising, e.g., one or more of TCRβ V5-6*01, TCRβ V5-4*01, or TCRβ V5-8*01;
    • (iv) TCRβ V12 subfamily comprising e.g., one or more of TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01;
    • (v) TCRβ V7 subfamily comprising e.g., one or more of TCRβ V7-7*01, TCRβ V7-6*01, TCRβ V7-8*02, TCRβ V7-4*01, TCRβ V7-2*02, TCRβ V7-2*03, TCRβ V7-2*01, TCRβ V7-3*01, TCRβ V7-9*03, or TCRβ V7-9*01;
    • (vi) TCRβ V11 subfamily comprising e.g., one or more of TCRβ V11-1*01, TCRβ V11-2*01 or TCRβ V11-3*01;
    • (vii) TCRβ V14 subfamily comprising TCRβ V14*01;
    • (viii) TCRβ V16 subfamily comprising TCRβ V16*01;
    • (ix) TCRβ V18 subfamily comprising TCRβ V18*01;
    • (x) TCRβ V9 subfamily comprising T e.g., one or more of CRβ V9*01 or TCRβ V9*02;
    • (xi) TCRβ V13 subfamily comprising TCRβ V13*01;
    • (xii) TCRβ V4 subfamily comprising e.g., one or more of e.g., one or more of TCRβ V4-2*01, TCRβ V4-3*01, or TCRβ V4-1*01;
    • (xiii) TCRβ V3 subfamily comprising TCRβ V3-1*01;
    • (xiv) TCRβ V2 subfamily comprising TCRβ V2*01;
    • (xv) TCRβ V15 subfamily comprising TCRβ V15*01;
    • (xvi) TCRβ V30 subfamily comprising e.g., one or more of TCRβ V30*01, or TCRβ V30*02;
    • (xvii) TCRβ V19 subfamily comprising e.g., one or more of TCRβ V19*01, or TCRβ V19*02;
    • (xviii) TCRβ V27 subfamily comprising TCRβ V27*01;
    • (xix) TCRβ V28 subfamily comprising TCRβ V28*01;
    • (xx) TCRβ V24 subfamily comprising TCRβ V24-1*01;
    • (xxi) TCRβ V20 subfamily comprising e.g., one or more of TCRβ V20-1*01, or TCRβ V20-1*02;
    • (xxii) TCRβ V25 subfamily comprising TCRβ V25-1*01; or
    • (xxiii) TCRβ V29 subfamily comprising TCRβ V29-1*01;
    • (xxiv) TCRβ V21 subfamily;
    • (xxv) TCRβ V1 subfamily;
    • (xxvi) TCRβ V17 subfamily;
    • (xvii) TCRβ V23 subfamily; or
    • (xviii) TCRβ V26 subfamily.

In some embodiments of any of the methods or composition for use disclosed herein, the cancer is a solid tumor including but not limited to: melanoma, pancreatic (e.g., pancreatic adenocarcinoma) cancer, breast cancer, colorectal cancer (CRC), lung cancer (e.g., small or non-small cell lung cancer), skin cancer, ovarian cancer, or liver cancer.

In some embodiments of any of the methods or composition for use disclosed herein, the cancer is a hematological cancer including, but not limited to: a B-cell or T cell malignancy, e.g., Hodgkin's lymphoma, Non-Hodgkin's lymphoma (e.g., B cell lymphoma, diffuse large B cell lymphoma (DLBCL), follicular lymphoma, chronic lymphocytic leukemia (B-CLL), mantle cell lymphoma, marginal zone B-cell lymphoma, Burkitt lymphoma, lymphoplasmacytic lymphoma, hairy cell leukemia), acute myeloid leukemia (AML), chronic myeloid leukemia, myelodysplastic syndrome, multiple myeloma, and acute lymphocytic leukemia.

In some embodiments of a method of expansion, or method of treatment, or composition for use disclosed herein, a higher, e.g., increased, level or activity of one or more TCRβV molecules in a subject, e.g., in a sample from a subject, is indicative of a bias, e.g., a preferential expansion, e.g., clonal expansion, of T cells expressing said one or more TCRβV molecules in the subject.

In some embodiments, a subject having a cancer, e.g., as disclosed herein, has a higher, e.g., increased, level or activity of one or more TCRβV molecules associated with the cancer. In some embodiments, the TCRβV molecule is associated with, e.g., recognizes, a cancer antigen, e.g., a cancer associated antigen or a neoantigen.

In some embodiments of any of the methods or composition for use disclosed herein, the subject has B-CLL. In some embodiments, a subject having B-CLL has a higher, e.g., increased, level or activity of one or more TCRβV molecules, e.g., one or more TCRβV molecules comprising: (i) TCRβ V6 subfamily comprising, e.g., TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01; (ii) TCRβ V5 subfamily comprising TCRβ V5-6*01, TCRβ V5-4*01, or TCRβ V5-8*01; (iii) TCRβ V3 subfamily comprising TCRβ V3-1*01; (iv) TCRβ V2 subfamily comprising TCRβ V2*01; or (v) TCRβ V19 subfamily comprising TCRβ V19*01, or TCRβ V19*02.

In some embodiments, a subject having B-CLL has a higher, e.g., increased, level or activity of a TCRβ V6 subfamily comprising, e.g., TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V6 subfamily. In some embodiments, administration of the an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V6 subfamily.

In some embodiments, a subject having B-CLL has a higher, e.g., increased, level or activity of a TCRβ V5 subfamily comprising TCRβ V5-6*01, TCRβ V5-4*01, or TCRβ V5-8*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V5 subfamily. In some embodiments, administration of the an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V5 subfamily.

In some embodiments, a subject having B-CLL has a higher, e.g., increased, level or activity of a TCRβ V3 subfamily comprising TCRβ V3-1*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V3 subfamily. In some embodiments, administration of an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V3 subfamily.

In some embodiments, a subject having B-CLL has a higher, e.g., increased, level or activity of a TCRβ V2 subfamily comprising TCRβ V2*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V2 subfamily. In some embodiments, administration of an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V2 subfamily.

In some embodiments, a subject having B-CLL has a higher, e.g., increased, level or activity of a TCRβ V19 subfamily comprising TCRβ V19*01, or TCRβ V19*02. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V19 subfamily. In some embodiments, administration of an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V19 subfamily.

In some embodiments of any of the methods or composition for use disclosed herein, the subject has melanoma. In some embodiments, a subject having melanoma has a higher, e.g., increased, level or activity of one or more TCRβV molecules, e.g., one or more TCRβV molecules comprising the TCRβ V6 subfamily comprising, e.g., TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V6 subfamily. In some embodiments, administration of an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V6 subfamily.

In some embodiments of any of the methods or composition for use disclosed herein, the subject has DLBCL. In some embodiments, a subject having melanoma has a higher, e.g., increased, level or activity of one or more TCRβV molecules, e.g., one or more TCRβV molecules comprising: (i) TCRβ V13 subfamily comprising TCRβ V13*01; (ii) TCRβ V3 subfamily comprising TCRβ V3-1*01; or (iii) TCRβ V23 subfamily.

In some embodiments, a subject having DLBCL has a higher, e.g., increased, level or activity of a TCRβ V13 subfamily comprising TCRβ V13*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V13 subfamily. In some embodiments, administration of an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V13 subfamily.

In some embodiments, a subject having DLBCL has a higher, e.g., increased, level or activity of a TCRβ V3 subfamily comprising TCRβ V3-1*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V3 subfamily. In some embodiments, administration of an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V3 subfamily.

In some embodiments, a subject having DLBCL has a higher, e.g., increased, level or activity of a TCRβ V23 subfamily. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V23 subfamily. In some embodiments, administration of an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V23 subfamily.

In some embodiments of any of the methods or composition for use disclosed herein, the subject has CRC. In some embodiments, a subject having melanoma has a higher, e.g., increased, level or activity of one or more TCRβV molecules, e.g., one or more TCRβV molecules comprising: (i) TCRβ V19 subfamily comprising TCRβ V19*01, or TCRβ V19*02; (ii) TCRβ V12 subfamily comprising TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01; (iii) TCRβ V16 subfamily comprising TCRβ V16*01; or (iv) TCRβ V21 subfamily.

In some embodiments, a subject having CRC has a higher, e.g., increased, level or activity of a TCRβ V19 subfamily comprising TCRβ V19*01, or TCRβ V19*02. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V19 subfamily. In some embodiments, administration of an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V19 subfamily.

In some embodiments, a subject having CRC has a higher, e.g., increased, level or activity of a TCRβ V12 subfamily comprising TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V12 subfamily. In some embodiments, administration of an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V12 subfamily.

In some embodiments, a subject having CRC has a higher, e.g., increased, level or activity of a TCRβ V16 subfamily comprising TCRβ V16*01. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V16 subfamily. In some embodiments, administration of an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V16 subfamily.

In some embodiments, a subject having CRC has a higher, e.g., increased, level or activity of a TCRβ V21 subfamily. In some embodiments, the subject is administered an anti-TCRβV molecule (e.g., an agonistic anti-TCRβV molecule as described herein) that binds to one or more members of the TCRβ V21 subfamily. In some embodiments, administration of an anti-TCRβV molecule results in expansion of immune cells expressing one or more members of the TCRβ V21 subfamily.

Alternatively or in combination with any of the embodiments disclosed herein, provided herein is an anti-TCRβV antibody molecule which:

    • (i) binds specifically to an epitope on TCRβV, e.g., the same or similar epitope as the epitope recognized by an anti-TCRβV antibody molecule as described herein, e.g., a second anti-TCRβV antibody molecule;
    • (ii) shows the same or similar binding affinity or specificity, or both, as an anti-TCRβV antibody molecule as described herein, e.g., a second anti-TCRβV antibody molecule;
    • (iii) inhibits, e.g., competitively inhibits, the binding of an anti-TCRβV antibody molecule as described herein, e.g., a second anti-TCRβV antibody molecule;
    • (iv) binds the same or an overlapping epitope with an anti-TCRβV antibody molecule as described herein, e.g., a second anti-TCRβV antibody molecule; or
    • (v) competes for binding, and/or binds the same epitope, with an anti-TCRβV antibody molecule as described herein, e.g., a second anti-TCRβV antibody molecule,

In some embodiments, the second anti-TCRβV antibody molecule comprises an antigen binding domain chosen from Table 1 or Table 2, or a sequence substantially identical thereto. In some embodiments, the second anti-TCRβV antibody molecule comprises an antigen binding domain, comprising:

a heavy chain complementarity determining region 1 (HC CDR1), a heavy chain complementarity determining region 2 (HC CDR2) and/or a heavy chain complementarity determining region 3 (HC CDR3) of SEQ ID NO: 1 or SEQ ID NO: 9; and/or a light chain complementarity determining region 1 (LC CDR1), a light chain complementarity determining region 2 (LC CDR2), and/or a light chain complementarity determining region 3 (LC CDR3) of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a heavy chain complementarity determining region 1 (HC CDR1), a heavy chain complementarity determining region 2 (HC CDR2) and/or a heavy chain complementarity determining region 3 (HC CDR3) of SEQ ID NO: 1 or SEQ ID NO: 9, or a sequence disclosed in Table 1; or
    • (ii) a light chain complementarity determining region 1 (LC CDR1), a light chain complementarity determining region 2 (LC CDR2), and/or a light chain complementarity determining region 3 (LC CDR3) of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11, or a sequence disclosed in Table 1.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of SEQ ID NO:1 or SEQ ID NO: 9.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a VL comprising: a LC CDR1 amino acid sequence of SEQ ID NO: 6 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), a LC CDR2 amino acid sequence of SEQ ID NO:7 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), and/or a LC CDR3 amino acid sequence of SEQ ID NO:8 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof); and/or
    • (ii) a VH comprising: a HC CDR1 amino acid sequence of SEQ ID NO: 3 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), a HC CDR2 amino acid sequence of SEQ ID NO:4 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), and/or a HC CDR3 amino acid sequence of SEQ ID NO:5 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof).

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • a variable heavy chain (VH) of SEQ ID NO: 9 or SEQ ID NO: 1312, or a sequence having at least about 75%, 80%, 85%, 90%, 95%, or 99% sequence identity thereto; and/or
    • a variable light chain (VL) of SEQ ID NO: 10 or SEQ ID NO: 11 or SEQ ID NO: 1314, or a sequence having at least about 75%, 80%, 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising the VH amino acid sequence of SEQ ID NO: 9 and the VL amino acid sequence of SEQ ID NO: 10.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising the VH amino acid sequence of SEQ ID NO: 9 and the VL amino acid sequence of SEQ ID NO: 11.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising the VH amino acid sequence of SEQ ID NO: 1312 and the VL amino acid sequence of SEQ ID NO: 1314.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising the amino acid sequence of SEQ ID NO: 1337, or a sequence with at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identity thereto.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising the amino acid sequence of SEQ ID NO: 1500, or a sequence with at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identity thereto.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises a heavy chain comprising a framework region, e.g., framework region 3 (FR3), comprising one or both of: (i) a Threonine at position 73, e.g., a substitution at position 73 according to Kabat numbering, e.g., a Glutamic Acid to Threonine substitution; or (ii) a Glycine at position, e.g., a substitution at position 94 according to Kabat numbering, e.g., a Arginine to Glycine substitution. In some embodiments, the substitution is relative to a human germline heavy chain framework region sequence.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises a light chain comprising a framework region, e.g., framework region 1 (FR1), comprising a Phenylalanine at position 10, e.g., a substitution at position 10 according to Kabat numbering, e.g., a Serine to Phenyalanine substitution. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises a light chain comprising a framework region, e.g., framework region 2 (FR2), comprising one or both of: (i) a Histidine at position 36, e.g., a substitution at position 36 according to Kabat numbering, e.g., a Tyrosine to Histidine substitution; or (ii) an Alanine at position 46, e.g., a substitution at position 46 according to Kabat numbering, e.g., a Arginine to Alanine substitution. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises a light chain comprising a framework region, e.g., framework region 3 (FR3), comprising a Phenylalanine at position 87, e.g., a substitution at position 87 according to Kabat numbering, e.g., a Tyrosine to Phenyalanine substitution. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule binds to TCRβ V6, e.g., TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01. In some embodiments the anti-TCRβV antibody molecule binds to TCRβ V6-5*01.

In some embodiments, TCRβ V6, e.g., TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01, is recognized, e.g., bound, by SEQ ID NO: 1 and/or SEQ ID NO: 2. In some embodiments, TCRβ V6, e.g., TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01, is recognized, e.g., bound, by SEQ ID NO: 9 and/or SEQ ID NO: 10. In some embodiments, TCRβ V6, e.g., TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01, is recognized, e.g., bound, by SEQ ID NO: 9 and/or SEQ ID NO: 11. In some embodiments, TCRβ V6-5*01 is recognized, e.g., bound by SEQ ID NO: 9 and/or SEQ ID NO: 10, or a sequence having at least about 75%, 80%, 85%, 90%, 95%, or 99% sequence identity thereto. In some embodiments, TCRβ V6-5*01 is recognized, e.g., bound by SEQ ID NO: 9 and/or SEQ ID NO: 11, or a sequence having at least about 75%, 80%, 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a heavy chain complementarity determining region (HC CDR1), a HC CDR2 and/or a HC CDR3 of SEQ ID NO: 15, SEQ ID NO: 23, SEQ ID NO: 24 or SEQ ID NO: 25, or a sequence disclosed in Table 2; and/or
    • (ii) a light chain complementarity determining region 1 (LC CDR1), a LC CDR2, and/or a LC CDR3 of SEQ ID NO: 16, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO:30, or a sequence disclosed in Table 2.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all of a LC CDR1, a LC CDR2 and a LC CDR3 of SEQ ID NO: 16, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO:30.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all of a HC CDR1, a HC CDR2 and a HC CDR3 of SEQ ID NO: 15, SEQ ID NO: 23, SEQ ID NO: 24 or SEQ ID NO: 25.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a VL comprising: a LC CDR1 amino acid sequence of SEQ ID NO: 20 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), a LC CDR2 amino acid sequence of SEQ ID NO:21 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), and/or a LC CDR3 amino acid sequence of SEQ ID NO:22 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof); and/or
    • (ii) a VH comprising: a HC CDR1 amino acid sequence of SEQ ID NO: 17 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), a HC CDR2 amino acid sequence of SEQ ID NO:18 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), and/or a HC CDR3 amino acid sequence of SEQ ID NO:19 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof).

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • a variable heavy chain (VH) of SEQ ID NO: 23, SEQ ID NO: 24 or SEQ ID NO: 25, or a sequence having at least about 75%, 80%, 85%, 90%, 95%, or 99% sequence identity thereto; and/or
    • a variable light chain (VL) of SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO:30, or a sequence having at least about 75%, 80%, 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises a light chain comprising a framework region, e.g., framework region 1 (FR1), comprising one, two or all (e.g., three) of: (i) an Aspartic Acid at position 1, e.g., a substitution at position 1 according to Kabat numbering, e.g., a Alanine to Aspartic Acid substitution; or (ii) an Asparagine at position 2, e.g., a substitution at position 2 according to Kabat numbering, e.g., a Isoleucine to Asparagine substitution, a Serine to Asparagine substitution, or a Tyrosine to Asparagine substitution; or (iii) a Leucine at position 4, e.g., a substitution at position 4 according to Kabat numbering, e.g., a Methionine to Leucine substitution. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises a light chain comprising a framework region, e.g., framework region 3 (FR3), comprising one, two or all (e.g., three) of: (i) a Glycine as position 66, e.g., a substitution at position 66 according to Kabat numbering, e.g., a Lysine to Glycine substitution, or a Serine to Glycine substitution; or (ii) an Asparagine at position 69, e.g., a substitution at position 69 according to Kabat numbering, e.g., a Threonine to Asparagine substitution; or (iii) a Tyrosine at position 71, e.g., a substitution at position 71 according to Kabat numbering, e.g., a Phenylalanine to Tyrosine substitution, or an Alanine to Tyrosine substitution. In some embodiments, the substitution is relative to a human germline light chain framework region sequence.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule binds to TCRβ V12, e.g., TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01. In some embodiments the anti-TCRβV antibody molecule binds to TCRβ V12-4*01 or TCRβ V12-3*01.

In some embodiments, TCRβ V12, e.g., TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01 is recognized, e.g., bound, by SEQ ID NO: 15 and/or SEQ ID NO: 16. In some embodiments, TCRβ V12, e.g., TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01, is recognized, e.g., bound, by any one of SEQ ID NOs 23-25, and/or any one of SEQ ID NO: 26-30, or an amino acid sequence having at least about 75%, 80%, 85%, 90%, 95%, or 99% sequence identity thereto. In some embodiments TCRβ V12-4*01 is recognized, e.g., bound, by any one of SEQ ID NOs 23-25, and/or any one of SEQ ID NO: 26-30, or an amino acid sequence having at least about 75%, 80%, 85%, 90%, 95%, or 99% sequence identity thereto. In some embodiments TCRβ V12-3*01 is recognized, e.g., bound, by any one of SEQ ID NOs 23-25, and/or any one of SEQ ID NO: 26-30, or an amino acid sequence having at least about 75%, 80%, 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a single chain Fv (scFv) or a Fab.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises binds to a conformational or a linear epitope on the T cell.

In some embodiments of any of the compositions or methods disclosed herein, the tumor comprises an antigen, e.g., a tumor antigen, e.g., a tumor associated antigen or a neoantigen. In some embodiments, the anti-TCRβV antibody molecule recognize, e.g., bind to, the tumor antigen.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule is a full antibody (e.g., an antibody that includes at least one, and preferably two, complete heavy chains, and at least one, and preferably two, complete light chains), or an antigen-binding fragment (e.g., a Fab, F(ab′)2, Fv, a single chain Fv fragment, a single domain antibody, a diabody (dAb), a bivalent antibody, or bispecific antibody or fragment thereof, a single domain variant thereof, a camelid antibody, or a rat-derived VH.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises the anti-TCRβV antibody molecule comprises one or more heavy chain constant regions chosen from IgG1, IgG2, IgG3, IgGA1, IgGA2, IgM, IgJ or IgG4, or a fragment thereof, e.g., as disclosed in Table 3.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises a heavy chain constant region of an IgM or a fragment thereof, optionally wherein the IgM heavy chain constant region comprises the sequence of SEQ ID NO: 73, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto. In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprising an IgM constant region, further comprises a heavy chain constant region of an IgJ or a fragment thereof, optionally wherein the IgJ heavy chain constant region comprises the sequence of SEQ ID NO: 76 or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises a heavy chain constant region of an IgJ or a fragment thereof, optionally wherein the IgJ heavy chain constant region comprises the sequence of SEQ ID NO: 76 or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises a heavy chain constant region of an IgGA1, or a fragment thereof, optionally wherein the IgGA1 heavy chain constant region comprises the sequence of SEQ ID NO: 74, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises a heavy chain constant region of an IgGA2, or a fragment thereof, optionally wherein the IgGA2 heavy chain constant region comprises a sequence listed in Table 3, e.g., SEQ ID NO: 75, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments of any of the compositions or methods disclosed herein, binding of the anti-TCRβV antibody molecule to a TCRβV region results in a cytokine profile, e.g., a cytokine secretion profile, (e.g., comprising one or more cytokines and/or one or more chemokines), that differs from that of a T cell engager that binds to a receptor or molecule other than a TCRβV region (“a non-TCRβV-binding T cell engager”).

In some embodiments, the cytokine profile, e.g., cytokine secretion profile, comprises the level and/or activity of one or more cytokines and/or one or more chemokines (e.g., as described herein). In an embodiment, a cytokine profile, e.g., a cytokine secretion profile, comprises the level and/or activity of one or more of: IL-2 (e.g., full length, a variant, or a fragment thereof); IL-1beta (e.g., full length, a variant, or a fragment thereof); IL-6 (e.g., full length, a variant, or a fragment thereof); TNFα (e.g., full length, a variant, or a fragment thereof); IFNg (e.g., full length, a variant, or a fragment thereof) IL-10 (e.g., full length, a variant, or a fragment thereof); IL-4 (e.g., full length, a variant, or a fragment thereof); TNF alpha (e.g., full length, a variant, or a fragment thereof); IL-12p70 (e.g., full length, a variant, or a fragment thereof); IL-13 (e.g., full length, a variant, or a fragment thereof); IL-8 (e.g., full length, a variant, or a fragment thereof); Eotaxin (e.g., full length, a variant, or a fragment thereof); Eotaxin-3 (e.g., full length, a variant, or a fragment thereof); IL-8 (HA) (e.g., full length, a variant, or a fragment thereof); IP-10 (e.g., full length, a variant, or a fragment thereof); MCP-1 (e.g., full length, a variant, or a fragment thereof); MCP-4 (e.g., full length, a variant, or a fragment thereof); MDC (e.g., full length, a variant, or a fragment thereof); MIP-1a (e.g., full length, a variant, or a fragment thereof); MIP-1b (e.g., full length, a variant, or a fragment thereof); TARC (e.g., full length, a variant, or a fragment thereof); GM-CSF (e.g., full length, a variant, or a fragment thereof); IL-12 23p40 (e.g., full length, a variant, or a fragment thereof); IL-15 (e.g., full length, a variant, or a fragment thereof); IL-16 (e.g., full length, a variant, or a fragment thereof); IL-17a (e.g., full length, a variant, or a fragment thereof); IL-1a (e.g., full length, a variant, or a fragment thereof); IL-5 (e.g., full length, a variant, or a fragment thereof); IL-7 (e.g., full length, a variant, or a fragment thereof); TNF-beta (e.g., full length, a variant, or a fragment thereof); or VEGF (e.g., full length, a variant, or a fragment thereof).

In some embodiments, the cytokine profile, e.g., cytokine secretion profile, comprises one, two, three, four, five, six, seven, or all of the following:

    • (i) increased level, e.g., expression level, and/or activity of IL-2;
    • (ii) reduced level, e.g., expression level, and/or activity of IL-1β;
    • (iii) reduced level, e.g., expression level, and/or activity of IL-6;
    • (iv) reduced level, e.g., expression level, and/or activity of TNFα;
    • (v) reduced level, e.g., expression level, and/or activity of IL-10;
    • (vi) a delay, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more hours delay, in increased level, e.g., expression level, and/or activity of IL-2;
    • (vii) a delay, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 hours delay, in increased level, e.g., expression level, and/or activity of IFNg; or
    • (viii) increased level, e.g., expression level, and/or activity of IL-15, e.g., wherein (i)-(viii) are relative to the cytokine profile, e.g., cytokine secretion profile, of the non-TCRβV-binding T cell engager.

In some embodiments, binding of the anti-TCRβV antibody to a TCRβV region results in reduced cytokine storm, e.g., reduced cytokine release syndrome (CRS) and/or neurotoxicity (NT), as measured by an assay of Example 3, e.g., relative to the cytokine storm induced by the non-TCRβV-binding T cell engager.

In some embodiments, binding of the anti-TCRβV antibody to a TCRβV region results in one, two, three or all of:

    • (ix) reduced T cell proliferation kinetics;
    • (x) cell killing, e.g., target cell killing, e.g. cancer cell killing, e.g., as measured by an assay of Example 4;
    • (xi) increased Natural Killer (NK) cell proliferation, e.g., expansion; or
    • (xii) expansion, e.g., at least about 1.1-10 fold expansion (e.g., at least about 1.1, 1.2, 1.3, 1.4, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, or 10 fold expansion), of a population of T cells having a memory-like phenotype,
    • e.g., wherein (ix)-(xii) are relative to the non-TCRβV-binding T cell engager.

In some embodiments, an anti-TCRβV antibody molecule disclosed herein recognizes (e.g., binds to), a structurally conserved domain on the TCRβV protein (e.g., as denoted by the circled area in FIG. 24A).

In some embodiments, an anti-TCRβV antibody molecule disclosed herein does not recognize, e.g., bind to, an interface of a TCRβV:TCRalpha complex.

In some embodiments, an anti-TCRβV antibody molecule disclosed herein does not recognize, e.g., bind to, a constant region of a TCRβV protein. An exemplary antibody that binds to a constant region of a TCRBV region is JOVI.1 as described in Viney et al., (Hybridoma. 1992 December; 11(6):701-13).

In some embodiments, an anti-TCRβV antibody molecule disclosed herein does not recognize, e.g., bind to, one or more (e.g., all) of a complementarity determining region (e.g., CDR1, CDR2 and/or CDR3) of a TCRβV protein.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises a light chain constant region chosen from the light chain constant regions of kappa or lambda, or a fragment thereof, e.g., as disclosed in Table 3.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises a light chain constant region of a kappa chain, or a fragment thereof, optionally wherein the kappa chain constant region comprises the sequence of SEQ ID NO: 39, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises:

    • (i) one or more heavy chain constant regions comprising a heavy chain constant region chosen from IgG1, IgG2, IgG3, IgGA1, IgGA2, IgG4, IgJ, IgM, IgD, or IgE, or a fragment thereof, e.g., as described in Table 3; and
    • (ii) a light chain constant region comprising a light chain constant region chosen from the light chain constant regions of kappa or lambda, or a fragment thereof, e.g., as described in Table 3.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises or a cell comprising an anti-TCRβV antibody molecule comprises:

    • (i) a heavy chain comprising a variable region (VH), e.g., a VH of an antibody disclosed herein; and/or one or more heavy chain constant regions, e.g., as disclosed herein; and/or
    • (ii) a light chain comprising a variable light (VL), e.g., a VL of an antibody disclosed herein; and/or one or more light chain constant regions, e.g., as disclosed herein.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises, or a cell comprising an anti-TCRβV antibody molecule comprises:

    • (i) a heavy chain comprising a heavy chain constant region comprising:
    • (a) an IgM heavy chain constant region or a fragment thereof, comprising the sequence of SEQ ID NO: 73, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto;
    • (b) an IgGA1 heavy chain constant region or a fragment thereof, comprising the sequence of SEQ ID NO: 74, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto; or
    • (c) an IgGA2 heavy chain constant region or a fragment thereof, comprising the sequence of SEQ ID NO: 75, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto; and
    • (ii) a light chain comprising a light chain constant region comprising a kappa chain constant region comprising the sequence of SEQ ID NO: 39, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto,
    • optionally wherein, the anti-TCRβV antibody molecule further comprises an IgJ heavy chain constant region or a fragment thereof, wherein the IgJ heavy chain constant region comprises the sequence of SEQ ID NO: 76 or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments of any of the compositions or methods disclosed herein, the anti-TCRβV antibody molecule comprises, or a cell comprising an anti-TCRβV antibody molecule comprises:

    • (i) a heavy chain comprising: a VH chosen from a VH of Tables 1-2 or 10-13, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto; and a heavy chain constant region comprising:
    • (a) an IgM heavy chain constant region or a fragment thereof, comprising the sequence of SEQ ID NO: 73, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto;
    • (b) an IgGA1 heavy chain constant region or a fragment thereof, comprising the sequence of SEQ ID NO: 74, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto; or
    • (c) an IgGA2 heavy chain constant region or a fragment thereof, comprising the sequence of SEQ ID NO: 75, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto; and
    • (ii) a light chain comprising: a VL chosen from a VL of Tables 1-2 or 10-13, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto; and a light chain constant region comprising a kappa chain constant region comprising the sequence of SEQ ID NO: 39, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto,
    • optionally wherein, the anti-TCRβV antibody molecule further comprises an IgJ heavy chain constant region or a fragment thereof, wherein the IgJ heavy chain constant region comprises the sequence of SEQ ID NO: 76 or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments of any of the methods disclosed herein, the anti-TCRβV antibody molecule binds to one or more (e.g., all) of the following TCRβV subfamilies:

    • (i) TCRβ V6 subfamily comprising, e.g., one or more of TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01;
    • (ii) TCRβ V10 subfamily comprising, e.g., one or more of TCRβ V10-1*01, TCRβ V10-1*02, TCRβ V10-3*01 or TCRβ V10-2*01;
    • (iii) TCRβ V5 subfamily comprising, e.g., one or more of TCRβ V5-6*01, TCRβ V5-4*01, or TCRβ V5-8*01;
    • (iv) TCRβ V12 subfamily comprising e.g., one or more of TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01;
    • (v) TCRβ V7 subfamily comprising e.g., one or more of TCRβ V7-7*01, TCRβ V7-6*01, TCRβ V7-8*02, TCRβ V7-4*01, TCRβ V7-2*02, TCRβ V7-2*03, TCRβ V7-2*01, TCRβ V7-3*01, TCRβ V7-9*03, or TCRβ V7-9*01;
    • (vi) TCRβ V11 subfamily comprising e.g., one or more of TCRβ V11-1*01, TCRβ V11-2*01 or TCRβ V11-3*01;
    • (vii) TCRβ V14 subfamily comprising TCRβ V14*01;
    • (viii) TCRβ V16 subfamily comprising TCRβ V16*01;
    • (ix) TCRβ V18 subfamily comprising TCRβ V18*01;
    • (x) TCRβ V9 subfamily comprising T e.g., one or more of CR(3 V9*01 or TCRβ V9*02;
    • (xi) TCRβ V13 subfamily comprising TCRβ V13*01;
    • (xii) TCRβ V4 subfamily comprising e.g., one or more of e.g., one or more of TCRβ V4-2*01, TCRβ V4-3*01, or TCRβ V4-1*01;
    • (xiii) TCRβ V3 subfamily comprising TCRβ V3-1*01;
    • (xiv) TCRβ V2 subfamily comprising TCRβ V2*01;
    • (xv) TCRβ V15 subfamily comprising TCRβ V15*01;
    • (xvi) TCRβ V30 subfamily comprising e.g., one or more of TCRβ V30*01, or TCRβ V30*02;
    • (xvii) TCRβ V19 subfamily comprising e.g., one or more of TCRβ V19*01, or TCRβ V19*02;
    • (xviii) TCRβ V27 subfamily comprising TCRβ V27*01;
    • (xix) TCRβ V28 subfamily comprising TCRβ V28*01;
    • (xx) TCRβ V24 subfamily comprising TCRβ V24-1*01;
    • (xxi) TCRβ V20 subfamily comprising e.g., one or more of TCRβ V20-1*01, or TCRβ V20-1*02;
    • (xxii) TCRβ V25 subfamily comprising TCRβ V25-1*01; or
    • (xxiii) TCRβ V29 subfamily comprising TCRβ V29-1*01;
    • (xxiv) TCRβ V21 subfamily;
    • (xxv) TCRβ V1 subfamily;
    • (xxvi) TCRβ V17 subfamily;
    • (xvii) TCRβ V23 subfamily; or
    • (xviii) TCRβ V26 subfamily.

In some embodiments of any of the methods disclosed herein, the anti-TCRβV antibody molecule binds to one or more (e.g., all) of the following TCRβV subfamilies:

    • (i) TCRβ V6, e.g., TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01;
    • (ii) TCRβ V10, e.g., TCRβ V10-1*01, TCRβ V10-1*02, TCRβ V10-3*01 or TCRβ V10-2*01;
    • (iii) TCRβ V12, e.g., TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01; or
    • (iv) TCRβ V5, e.g., TCRβ V5-5*01, TCRβ V5-6*01, TCRβ V5-4*01, TCRβ V5-8*01, TCRβ V5-1*01.

In some embodiments, the anti-TCRβV antibody molecule binds to TCRβ V6, e.g., TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01. In some embodiments, the anti-TCRβV antibody molecule binds to TCRβ V6-5*01.

In some embodiments, the anti-TCRβV antibody molecule does not bind to TCRβ V12.

In some embodiments, the anti-TCRβV antibody molecule does not bind to TCRβ V5-5*01 or TCRβ V5-1*01.

In some embodiments of any of the methods disclosed herein, the anti-TCRβV antibody molecule does not bind to TCRβ V12, or binds to TCRβ V12 with an affinity and/or binding specificity that is less than (e.g., less than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the 16G8 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments of any of the methods disclosed herein, the anti-TCRβV antibody molecule binds to TCRβ V12 with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the 16G8 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments of any of the methods disclosed herein, the anti-TCRβV antibody molecule binds to a TCRβV region other than TCRβ V12 (e.g., TCRβV region as described herein, e.g., TCRβ V6 subfamily (e.g., TCRβ V6-5*01) with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the 16G8 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments of any of the methods disclosed herein, the anti-TCRβV antibody molecule does not comprise at least one CDR of Antibody B. In some embodiments of any of the methods disclosed herein, the anti-TCRβV antibody molecule does not comprise the CDRs of Antibody B.

In some embodiments of any of the methods disclosed herein, the anti-TCRβV antibody molecule does not bind to TCRβ V5-5*01 or TCRβ V5-1*01, or binds to TCRβ V5-5*01 or TCRβ V5-1*01 with an affinity and/or binding specificity that is less than (e.g., less than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the TM23 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments of any of the methods disclosed herein, the anti-TCRβV antibody molecule binds to TCRβ V5-5*01 or TCRβ V5-1*01 with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the TM23 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments of any of the methods disclosed herein, the anti-TCRβV antibody molecule binds to a TCRβV region other than TCRβ V5-5*01 or TCRβ V5-1*01 (e.g., TCRβV region as described herein, e.g., TCRβ V6 subfamily (e.g., TCRβ V6-5*01) with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the TM23 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155.

In some embodiments of any of the methods disclosed herein, the anti-TCRβV antibody molecule does not comprise at least one CDR of the TM23 murine antibody. In some embodiments of any of the methods disclosed herein, the anti-TCRβV antibody molecule does not comprise the CDRs of the TM23 murine antibody.

In some embodiments of any of the methods disclosed herein, an anti-TCRβV antibody molecule disclosed herein does not comprise the sequence of a murine anti-rat TCR antibody R73, e.g., as disclosed in J Exp Med. 1989 Jan. 1; 169(1): 73-86, herein incorporated by reference in its entirety. In some embodiments of any of the methods disclosed herein, a multispecific antibody molecule disclosed herein does not comprise the sequence of a murine anti-rat TCR antibody R73, e.g., as disclosed in J Immunol. 1993 Mar. 15; 150(6):2305-15, herein incorporated by reference in its entirety.

In some embodiments of any of the methods disclosed herein, an anti-TCRβV antibody molecule disclosed herein does not comprise a viral peptide-WIC complex, e.g., as disclosed in Oncoimmunology. 2016; 5(1): e1052930, herein incorporated by reference in its entirety. In some embodiments of any of the methods disclosed herein, a multispecific antibody molecule disclosed herein does not comprise a viral peptide-WIC complex, e.g., as disclosed in Oncoimmunology. 2016; 5(1): e1052930, herein incorporated by reference in its entirety.

In some embodiments of a method disclosed herein, the immune cell population comprises a T cell, a Natural Killer cell, a B cell, an antigen presenting cell, or a myeloid cell (e.g., a monocyte, a macrophage, a neutrophil or a granulocyte).

In some embodiments of a method disclosed herein, the immune cell population comprises a T cell, e.g., a CD4+ T cell, a CD8+ T cell, a TCR alpha-beta T cell, or a TCR gamma-delta T cell. In some embodiments, a T cell comprises a memory T cell (e.g., a central memory T cell, or an effector memory T cell (e.g., a TEMRA) or an effector T cell. In some embodiments, a T cell comprises a tumor infiltrating lymphocyte (TIL).

In some embodiments of a method disclosed herein, the immune cell population is obtained from a healthy subject.

In some embodiments of a method disclosed herein, the immune cell population is obtained from a subject (e.g., from an apheresis sample from the subject) having a disease, e.g., a cancer, e.g., as described herein. In some embodiments, the immune cell population obtained from a subject having a disease, e.g., a cancer, comprises a tumor infiltrating lymphocyte (TIL).

In some embodiments of a method disclosed herein, the method results in an expansion of at least 1.1-10 fold (e.g., at least 1.1, 1.2, 1.3, 1.4, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, or 10 fold expansion).

In some embodiments of a method disclosed herein, the method further comprises contacting the population of cells with an agent that promotes, e.g., increases, immune cell expansion. In some embodiments, the agent includes an immune checkpoint inhibitor, e.g., as described herein. In some embodiments, the agent includes a 4-1BB (CD127) agonist, e.g., an anti-4-1BB antibody.

In some embodiments of a method disclosed herein, the method further comprises comprising contacting the population of cells with a non-dividing population of cells, e.g., feeder cells, e.g., irradiated allogenic human PBMCs.

In some embodiments of a method disclosed herein, an expansion method described herein comprises expanding the cells for a period of at least about 4 hours, 6 hours, 10 hours, 12 hours, 15 hours, 18 hours, 20 hours, or 22 hours, or for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 1,6 17, 18, 19, 20 or 21 days, or for at least about 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks or 8 weeks.

In some embodiments of a method disclosed herein, expansion of the population of immune cells, is compared to expansion of a similar population of cells with an antibody that binds to: a CD3 molecule, e.g., CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments of a method disclosed herein, expansion of the population of immune cells, is compared to expansion of a similar population of cells not contacted with the anti-TCRβV antibody molecule.

In some embodiments of a method disclosed herein, expansion of the population of memory effector T cells, e.g., TEM cells, e.g., TEMRA cells, is compared to expansion of a similar population of cells with an antibody that binds to: a CD3 molecule, e.g., CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments of a method disclosed herein, the method results in expansion of, e.g., selective or preferential expansion of, T cells expressing a T cell receptor (TCR) comprising a TCR alpha and/or TCR beta molecule, e.g., TCR alpha-beta T cells (αβ T cells).

In some embodiments of a method disclosed herein, the method results in expansion of αβT cells over expansion of T cells expressing a TCR comprising a TCR gamma and/or TCR delta molecule, e.g., TCR gamma-delta T cells (γδ T cells). In some embodiments, expansion of αβT cells over γδ T cells results in reduced production of cytokines associated with CRS. In some embodiments, expansion of αβT cells over γδ T cells results in immune cells that have reduced capacity to, e.g., are less prone to, induce CRS upon administration into a subject.

In some embodiments of a method disclosed herein, an immune cell population (e.g., T cells (e.g., TEMRA cells or TILs) or NK cells) cultured in the presence of, e.g., expanded with, an anti-TCRβV antibody disclosed herein does not induce CRS and/or NT when administered into a subject, e.g., a subject having a disease or condition as described herein.

In some embodiments, the anti-TCRβV antibody molecule in a multispecific molecule disclosed herein is a first immune cell engager moiety. In some embodiments, the anti-TCRβV antibody molecule does not bind to TCRβ V12, or binds to TCRβ V12 with an affinity and/or binding specificity that is less than (e.g., less than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the 16G8 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155. In some embodiments, the anti-TCRβV antibody molecule binds to TCRβ V12 with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the 16G8 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155. In some embodiments, the anti-TCRβV antibody molecule binds to a TCRβV region other than TCRβ V12 (e.g., TCRβV region as described herein, e.g., TCRβ V6 subfamily (e.g., TCRβ V6-5*01) with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the 16G8 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155. In some embodiments, the anti-TCRβV antibody molecule does not comprise the CDRs of the Antibody B murine antibody.

In some embodiments, the anti-TCRβV antibody molecule in a multispecific molecule disclosed herein is a first immune cell engager moiety. In some embodiments, the anti-TCRβV antibody molecule does not bind to TCRβ V5-5*01 or TCRβ V5-1*01, or binds to TCRβ V5-5*01 or TCRβ V5-1*01 with an affinity and/or binding specificity that is less than (e.g., less than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the TM23 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155. In some embodiments, the anti-TCRβV antibody molecule binds to TCRβ V5-5*01 or TCRβ V5-1*01 with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the TM23 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155. In some embodiments, the anti-TCRβV antibody molecule binds to a TCRβV region other than TCRβ V5-5*01 or TCRβ V5-1*01 (e.g., TCRβV region as described herein, e.g., TCRβ V6 subfamily (e.g., TCRβ V6-5*01) with an affinity and/or binding specificity that is greater than (e.g., greater than about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or about 2-, 5-, or 10-fold) the affinity and/or binding specificity of the TM23 murine antibody or a humanized version thereof as described in U.S. Pat. No. 5,861,155. In some embodiments, the anti-TCRβV antibody molecule does not comprise the CDRs of the TM23 murine antibody.

In some embodiments, the multispecific molecule further comprises a second immune cell engager moiety. In some embodiments, the first and/or second immune cell engager binds to and activates an immune cell, e.g., an effector cell. In some embodiments, the first and/or second immune cell engager binds to, but does not activate, an immune cell, e.g., an effector cell. In some embodiments, the second immune cell engager is chosen from an NK cell engager, a T cell engager, a B cell engager, a dendritic cell engager, or a macrophage cell engager, or a combination thereof. In some embodiments, the second immune cell engager comprises a T cell engager which binds to CD3, TCRα, TCRγ, TCRζ, ICOS, CD28, CD27, HVEM, LIGHT, CD40, 4-1BB, OX40, DR3, GITR, CD30, TIM1, SLAM, CD2, or CD226.

In some embodiments, a multispecific molecule disclosed herein comprises a tumor targeting moiety. In some embodiment, the tumor-targeting moiety comprises an antibody molecule (e.g., Fab or scFv), a receptor molecule (e.g., a receptor, a receptor fragment or functional variant thereof), or a ligand molecule (e.g., a ligand, a ligand fragment or functional variant thereof), or a combination thereof, that binds to a cancer antigen. In some embodiments, the tumor-targeting moiety binds to a cancer antigen present on a cancer, e.g., a hematological cancer, a solid tumor, a metastatic cancer, soft tissue tumor, metastatic lesion, or a combination thereof. In some embodiments, the tumor-targeting moiety binds to a cancer antigen, e.g., BCMA or FcRH5.

In some embodiments, the tumor-targeting antibody molecule binds to a conformational or a linear epitope on the tumor antigen.

In some embodiments of any of the compositions or methods disclosed herein, the tumor-targeting moiety is an antigen, e.g., a cancer antigen. In some embodiments, the cancer antigen is a tumor antigen or stromal antigen, or a hematological antigen.

In some embodiments of any of the compositions or methods disclosed herein, the tumor-targeting moiety binds to a cancer antigen chosen from: BCMA, FcRH5, CD19, CD20, CD22, CD30, CD33, CD38, CD47, CD99, CD123, FcRH5, CLEC12, CD179A, SLAMF7, or NY-ESO1, PDL1, CD47, gangloside 2 (GD2), prostate stem cell antigen (PSCA), prostate specific membrane antigen (PMSA), prostate-specific antigen (PSA), carcinoembryonic antigen (CEA), Ron Kinase, c-Met, Immature laminin receptor, TAG-72, BING-4, Calcium-activated chloride channel 2, Cyclin-B1, 9D7, Ep-CAM, EphA3, Her2/neu, Telomerase, SAP-1, Survivin, NY-ESO-1/LAGE-1, PRAME, SSX-2, Melan-A/MART-1, Gp100/pme117, Tyrosinase, TRP-1/-2, MC1R, β-catenin, BRCA1/2, CDK4, CML66, Fibronectin, p53, Ras, TGF-B receptor, AFP, ETA, MAGE, MUC-1, CA-125, BAGE, GAGE, NY-ESO-1, β-catenin, CDK4, CDC27, α actinin-4, TRP1/gp75, TRP2, gp100, Melan-A/MART1, gangliosides, WT1, EphA3, Epidermal growth factor receptor (EGFR), MART-2, MART-1, MUC1, MUC2, MUM1, MUM2, MUM3, NA88-1, NPM, OA1, OGT, RCC, RUI1, RUI2, SAGE, TRG, TRP1, TSTA, Folate receptor alpha, L1-CAM, CAIX, gpA33, GD3, GM2, VEGFR, Integrins (Integrin alphaVbeta3, Integrin alpha5Beta1), Carbohydrates (Le), IGF1R, EPHA3, TRAILR1, TRAILR2, RANKL, (FAP), TGF-beta, hyaluronic acid, collagen, e.g., collagen IV, tenascin C, or tenascin W.

In some embodiments of any of the compositions or methods disclosed herein, the cancer is a solid tumor including but not limited to: pancreatic (e.g., pancreatic adenocarcinoma) cancer, breast cancer, colorectal cancer, lung cancer (e.g., small or non-small cell lung cancer), skin cancer, ovarian cancer, or liver cancer.

In some embodiments of any of the compositions or methods disclosed herein, the cancer antigen or tumor antigen is a hematological antigen. In some embodiments, the cancer or tumor antigen is chosen from one or more of: BCMA, FcRH5, CD19, CD20, CD22, CD30, CD33, CD38, CD47, CD99, CD123, FcRH5, CLEC12, CD179A, SLAMF7, or NY-ESO1. In some embodiments, the tumor-targeting moiety binds to one or both of BCMA or FcRH5.

In some embodiments, the tumor-targeting moiety binds to BCMA. In embodiments, the tumor-targeting moiety comprises a BCMA targeting moiety. In some embodiments, the tumor-targeting moiety comprising a BCMA targeting moiety binds to a BCMA antigen on the surface of a cell, e.g., a cancer or hematopoietic cell. The BCMA antigen can be present on a primary tumor cell, or a metastatic lesion thereof. In some embodiments, the cancer is a hematological cancer, e.g., multiple myeloma. For example, the BCMA antigen can be present on a tumor, e.g., a tumor of a class typified by having one or more of: limited tumor perfusion, compressed blood vessels, or fibrotic tumor interstitium. In some embodiments, the tumor targeting moiety comprising a BCMA targeting moiety comprises an anti-BCMA antibody or antigen-binding fragment thereof described in U.S. Pat. Nos. 8,920,776, 9,243,058, 9,340,621, 8,846,042, 7,083,785, 9,545,086, 7,276,241, 9,034,324, 7,799,902, 9,387,237, 8,821,883, US861745, US20130273055, US20160176973, US20150368351, US20150376287, US20170022284, US20160015749, US20140242077, US20170037128, US20170051068, US20160368988, US20160311915, US20160131654, US20120213768, US20110177093, US20160297885, EP3137500, EP2699259, EP2982694, EP3029068, EP3023437, WO2016090327, WO2017021450, WO2016110584, WO2016118641, WO2016168149, the entire contents of which are incorporated herein by reference.

In some embodiments, the BCMA-targeting moiety includes an antibody molecule (e.g., Fab or scFv) that binds to BCMA. In some embodiments, the antibody molecule to BCMA comprises one, two, or three CDRs from any of the heavy chain variable domain sequences of Table 14, or a closely related CDR, e.g., CDRs which have at least one amino acid alteration, but not more than two, three or four alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions) from any of the CDR sequences of Table 14. In some embodiments, the antibody molecule to BCMA comprises a heavy chain variable domain sequence chosen from any of the amino acid sequences of Table 14, or an amino acid sequence substantially identical thereto (e.g., 95% to 99.9% identical thereto, or having at least one amino acid alteration, but not more than five, ten or fifteen alterations (e.g., substitutions, deletions, or insertions, e.g., conservative substitutions)).

In some embodiments, the tumor-targeting moiety binds to FcRH5. In embodiments, the tumor-targeting moiety comprises a FcRH5targeting moiety. In some embodiments, the tumor-targeting moiety comprising a FcRH5targeting moiety binds to a FcRH5antigen on the surface of a cell, e.g., a cancer or hematopoietic cell. The FcRH5antigen can be present on a primary tumor cell, or a metastatic lesion thereof. In some embodiments, the cancer is a hematological cancer, e.g., multiple myeloma. For example, the FcRH5antigen can be present on a tumor, e.g., a tumor of a class typified by having one or more of: limited tumor perfusion, compressed blood vessels, or fibrotic tumor interstitium. In some embodiments, the tumor targeting moiety comprising a FcRH5targeting moiety comprises an anti-FcRH5antibody or antigen-binding fragment thereof described in U.S. Pat. No. 7,999,077 the entire contents of which are incorporated herein by reference.

In some embodiments of any of the compositions or methods disclosed herein, the cancer is a hematological cancer including, but not limited to: a B-cell or T cell malignancy, e.g., Hodgkin's lymphoma, Non-Hodgkin's lymphoma (e.g., B cell lymphoma, diffuse large B cell lymphoma, follicular lymphoma, chronic lymphocytic leukemia, mantle cell lymphoma, marginal zone B-cell lymphoma, Burkitt lymphoma, lymphoplasmacytic lymphoma, hairy cell leukemia), acute myeloid leukemia (AML), chronic myeloid leukemia, myelodysplastic syndrome, multiple myeloma, and acute lymphocytic leukemia. In some embodiments, the hematological cancer is multiple myeloma.

In some embodiments, a multispecific molecule disclosed herein further comprises a cytokine molecule, e.g., one or two cytokine molecules. In some embodiments, the cytokine molecule is chosen from interleukin-2 (IL-2), interleukin-7 (IL-7), interleukin-12 (IL-12), interleukin-15 (IL-15), interleukin-18 (IL-18), interleukin-21 (IL-21), or interferon gamma, or a fragment, variant or combination thereof. In some embodiments, is a monomer or a dimer. In some embodiments, the cytokine molecule further comprises a receptor dimerizing domain, e.g., an IL15Ralpha dimerizing domain. In some embodiments, the cytokine molecule (e.g., IL-15) and the receptor dimerizing domain (e.g., an IL15Ralpha dimerizing domain) are not covalently linked, e.g., are non-covalently associated.

In some embodiments, a multispecific molecule disclosed herein comprises:

    • (i) an anti-TCRβV antibody molecule (e.g., an anti-TCRβV antibody molecule as described herein); and
    • (ii) a tumor-targeting antibody molecule (e.g., an antibody molecule that binds to a hematological antigen as described herein, e.g., chosen from one or more of BCMA, FcRH5, CD19, CD22, CD33, CD123, FcRH5, CD179a, or CLEC12).

In some embodiments, the multispecific molecule disclosed herein comprises the anti-TCRβV antibody molecule of (i), the tumor-targeting antibody molecule of (ii) and a cytokine molecule as described herein, e.g., an IL-12 cytokine molecule.

In some embodiments, the multispecific molecule comprises an anti-TCRβV antibody molecule as described herein; and a tumor-targeting antibody molecule that binds to one or both of BCMA or FcRH5. In some embodiments, the multispecific molecule further comprises an IL-12 cytokine molecule. The multispecific molecule can be used to treat a BCMA- or FcRH5-expressing hematological cancer, e.g., multiple myeloma.

In some embodiments, the multispecific molecule comprises an anti-TCRβV antibody molecule as described herein; and a tumor-targeting antibody molecule that binds one or more of CD19, CD22, or CD123. In some embodiments, the multispecific molecule further comprises an IL-12 cytokine molecule. The multispecific molecule can be used to treat a CD19-, CD22-, or CD123-expressing hematological cancer, e.g., leukemia or lymphoma. In some embodiments, the CD19-, CD22-, or CD123-expressing hematological cancer is chosen from a B-cell or T cell malignancy, e.g., Hodgkin's lymphoma, Non-Hodgkin's lymphoma (e.g., B cell lymphoma, diffuse large B cell lymphoma, follicular lymphoma, chronic lymphocytic leukemia, mantle cell lymphoma, marginal zone B-cell lymphoma, Burkitt lymphoma, lymphoplasmacytic lymphoma, hairy cell leukemia), acute myeloid leukemia (AML), chronic myeloid leukemia, myelodysplastic syndrome, multiple myeloma, and acute lymphocytic leukemia. In some embodiments, the hematological cancer is multiple myeloma.

In some embodiments, a multispecific molecule disclosed herein further comprises an immunoglobulin constant region (e.g., Fc region) chosen from the heavy chain constant regions of IgG1, IgG2, and IgG4, more particularly, the heavy chain constant region of human IgG1, IgG2 or IgG4. In some embodiments, the immunoglobulin constant region (e.g., an Fc region) is linked, e.g., covalently linked to, one or more of tumor-targeting moiety, the immune cell engager, the cytokine molecule, or the stromal modifying moiety. In some embodiments, an interface of a first and second immunoglobulin chain constant regions (e.g., Fc region) is altered, e.g., mutated, to increase or decrease dimerization, e.g., relative to a non-engineered interface. In some embodiments, the dimerization of the immunoglobulin chain constant region (e.g., Fc region) is enhanced by providing an Fc interface of a first and a second Fc region with one or more of: a paired cavity-protuberance (“knob-in-a hole”), an electrostatic interaction, or a strand-exchange, such that a greater ratio of heteromultimer:homomultimer forms, e.g., relative to a non-engineered interface. In some embodiments,

In some embodiments, a multispecific molecule disclosed herein further comprises a linker, e.g., a linker described herein, optionally wherein the linker is selected from: a cleavable linker, a non-cleavable linker, a peptide linker, a flexible linker, a rigid linker, a helical linker, or a non-helical linker.

In some embodiments, the multispecific molecule comprises at least two non-contiguous polypeptide chains.

In some embodiments, the multispecific molecule comprises the following configuration:

    • A, B-[dimerization module]-C, -D
    • wherein:
    • (1) the dimerization module comprises an immunoglobulin constant domain, e.g., a heavy chain constant domain (e.g., a homodimeric or heterodimeric heavy chain constant region, e.g., an Fc region), or a constant domain of an immunoglobulin variable region (e.g., a Fab region); and
    • (2) A, B, C, and D are independently absent; (i) an antigen binding domain that preferentially binds to a first immune cell engager comprising an anti-TCRβV antibody molecule disclosed herein; (ii) a tumor targeting moiety (e.g., a tumor-targeting antibody molecule as described herein), (iii) a second immune cell engager chosen from a T cell engager, an NK cell engager, a B cell engager, a dendritic cell engager, or a macrophage cell engager; (iv) a cytokine molecule; or (v) a stromal modifying moiety, provided that:
    • at least one, two, or three of A, B, C, and D comprises an antigen binding domain that preferentially binds to a TCRβV region disclosed herein, and
    • any of the remaining A, B, C, and D is absent or comprises one of a tumor targeting moiety, a second immune cell engager, a cytokine molecule, or a stromal modifying moiety.

In some embodiments, the dimerization module comprises one or more immunoglobulin chain constant regions (e.g., Fc regions) comprising one or more of: a paired cavity-protuberance (“knob-in-a hole”), an electrostatic interaction, or a strand-exchange. In some embodiments, the one or more immunoglobulin chain constant regions (e.g., Fc regions) comprise an amino acid substitution at a position chosen from one or more of 347, 349, 350, 351, 366, 368, 370, 392, 394, 395, 397, 398, 399, 405, 407, or 409, e.g., of the Fc region of human IgG1. In some embodiments, the one or more immunoglobulin chain constant regions (e.g., Fc regions) comprise an amino acid substitution chosen from: T366S, L368A, or Y407V (e.g., corresponding to a cavity or hole), or T366W (e.g., corresponding to a protuberance or knob), or a combination thereof.

In some embodiments, the multispecific molecule further comprises a linker, e.g., a linker between one or more of: the antigen binding domain of an anti-TCRβV antibody molecule disclosed herein and the tumor targeting moiety; the antigen binding domain of an anti-TCRβV antibody molecule disclosed herein and the second immune cell engager, the antigen binding domain of an anti-TCRβV antibody molecule disclosed herein and the cytokine molecule, the antigen binding domain of an anti-TCRβV antibody molecule disclosed herein and the stromal modifying moiety, the second immune cell engager and the cytokine molecule, the second immune cell engager and the stromal modifying moiety, the cytokine molecule and the stromal modifying moiety, the antigen binding domain of an anti-TCRβV antibody molecule disclosed herein and the dimerization module, the second immune cell engager and the dimerization module, the cytokine molecule and the dimerization module, the stromal modifying moiety and the dimerization module, the tumor targeting moiety and the dimerization module, the tumor targeting moiety and the cytokine molecule, the tumor targeting moiety and the second immune cell engager, or the tumor targeting moiety and the antigen binding domain of an anti-TCRβV antibody molecule disclosed herein. In some embodiments, the linker is chosen from: a cleavable linker, a non-cleavable linker, a peptide linker, a flexible linker, a rigid linker, a helical linker, or a non-helical linker. In some embodiments, the linker is a peptide linker. In some embodiments, the peptide linker comprises Gly and Ser. In some embodiments, the peptide linker comprises an amino acid sequence chosen from SEQ ID NOs: 142-145 or 175-178.

In some embodiments of a method or composition for use disclosed herein, the disease is a cancer chosen from: a hematological cancer, a solid tumor, a metastatic cancer, soft tissue tumor, metastatic lesion, or a combination thereof.

In some embodiments of a method or composition for use disclosed herein, the cancer is a solid tumor chosen from: a melanoma, a pancreatic cancer (e.g., pancreatic adenocarcinoma), a breast cancer, a colorectal cancer (CRC), a lung cancer (e.g., small or non-small cell lung cancer), a skin cancer, an ovarian cancer, or a liver cancer. In some embodiments, the cancer is melanoma or CRC.

In some embodiments of a method or composition for use disclosed herein the cancer is a hematological cancer chosen from: a B-cell or T cell malignancy, e.g., Hodgkin's lymphoma, Non-Hodgkin's lymphoma (e.g., B cell lymphoma, diffuse large B cell lymphoma, follicular lymphoma, chronic lymphocytic leukemia, mantle cell lymphoma, marginal zone B-cell lymphoma, Burkitt lymphoma, lymphoplasmacytic lymphoma, hairy cell leukemia), acute myeloid leukemia (AML), chronic myeloid leukemia, myelodysplastic syndrome, multiple myeloma, or acute lymphocytic leukemia. In some embodiments, the hematological cancer is multiple myeloma. In some embodiments, the hematological cancer is CLL or DLBCL.

In some embodiments of a method or composition for use disclosed herein the sample from the subject comprises a blood sample, e.g., a peripheral blood sample, a biopsy, e.g., a tumor biopsy, or a bone marrow sample. In some embodiments, the sample comprises a biological sample comprising immune effector cells, e.g., T cells, or NK cells. In some embodiments, T cells comprise a CD4 T cell, a CD8 T cell, (e.g., an effector T cell or a memory T cell (e.g., a memory effector T cell (e.g., TEM cell, e.g., TEMRA cell), or a tumor infiltrating lymphocyte (TIL).

In some aspects, provided herein is, inter alia, a multispecific molecule (e.g., a bispecific molecule), comprising a first moiety (e.g., a first immune cell engager) comprising an antibody molecule which binds (e.g., specifically binds) to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”), wherein binding of the first moiety to the TCRβV region results in a cytokine profile that differs from a cytokine profile of a T cell engager that binds to a receptor or molecule other than a TCRβV region (“a non-TCRβV-binding T cell engager”).

In some embodiments, the multispecific molecule as provided herein comprises a second moiety which comprises one or more of: a tumor-targeting moiety, a cytokine molecule, a stromal modifying moiety, or an anti-TCRβV antibody molecule other than the first moiety.

In some embodiments, the first moiety comprising the anti-TCRβV antibody molecule comprises an Fc region comprising a variant, e.g., an Fc variant described in Table 21, e.g., an Asn297Ala (N297A) mutation or a Leu234A1a/Leu235Ala (LALA) mutation.

In some embodiments, the non-TCRβV-binding T cell engager comprises an antibody that binds to a CD3 molecule (e.g., CD3 epsilon (CD3e) molecule); or a TCR alpha (TCRα) molecule.

In some embodiments, the cytokine profile of the first moiety comprises, one, two, three, four, five, six, seven, or all of the following:

    • (i) increased level, e.g., expression level, and/or activity of IL-2;
    • (ii) reduced level, e.g., expression level, and/or activity of IL-1β;
    • (iii) reduced level, e.g., expression level, and/or activity of IL-6;
    • (iv) reduced level, e.g., expression level, and/or activity of TNFα;
    • (v) reduced level, e.g., expression level, and/or activity of IL-10;
    • (vi) a delay, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more hours delay, in increased level, e.g., expression level, and/or activity of IL-2;
    • (vii) a delay, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 hours delay, in increased level, e.g., expression level, and/or activity of IFNg; or
    • (viii) increased level, e.g., expression level, and/or activity of IL-15, e.g., wherein (i)-(viii) are relative to the cytokine profile of the non-TCRβV-binding T cell engager.

In some embodiments, binding of the first moiety to the TCRβV region results in reduced cytokine storm, e.g., reduced cytokine release syndrome (CRS), as measured by an assay of Example 3, e.g., relative to the cytokine storm induced by the non-TCRβV-binding T cell engager.

In some embodiments, binding of the first moiety to the TCRβV region results in one, two, three or all of:

    • (ix) reduced T cell proliferation kinetics;
    • (x) cell killing, e.g., target cell killing, e.g. cancer cell killing, e.g., as measured by an assay of Example 4;
    • (xi) increased Natural Killer (NK) cell proliferation, e.g., expansion; or
    • (xii) expansion, e.g., at least about 1.1-10 fold expansion (e.g., at least about 1.1, 1.2, 1.3, 1.4, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, or 10 fold expansion), of a population of T cells having a memory-like phenotype, e.g., wherein (ix)-(xii) are relative to the non-TCRβV-binding T cell engager.

In some embodiments, the population of T cells having a memory-like phenotype comprises CD45RA+ CCR7− T cells, e.g., CD4+ and/or CD8+ T cells.

In some embodiments, the first moiety binds to one or more of a TCRβV subfamily chosen from:

    • (i) TCRβ V6 subfamily comprising, e.g., one or more of TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01;
    • (ii) TCRβ V10 subfamily comprising, e.g., one or more of TCRβ V10-1*01, TCRβ V10-1*02, TCRβ V10-3*01 or TCRβ V10-2*01;
    • (iii) TCRβ V5 subfamily comprising, e.g., one or more of TCRβ V5-6*01, TCRβ V5-4*01, TCRβ V5-1*01 or TCRβ V5-8*01;
    • (iv) TCRβ V12 subfamily comprising, e.g., one or more of TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01;
    • (v) TCRβ V27 subfamily;
    • (vi) TCRβ V28 subfamily;
    • (vii) TCRβ V4 subfamily comprising, e.g., one or more of TCRβ V4-1, TCRβ V4-2 or TCRβ V4-3;
    • (viii) TCRβ V19 subfamily;
    • (ix) TCRβ V9 subfamily; or
    • (x) TCRβ V11 subfamily comprising, e.g., TCRβ V11-2.

In some embodiments, the anti-TCRβV antibody molecule:

    • (i) binds specifically to an epitope on TCRβV, e.g., the same or similar epitope as the epitope recognized by an anti-TCRβV antibody molecule as described herein, e.g., a second anti-TCRβV antibody molecule;
    • (ii) shows the same or similar binding affinity or specificity, or both, as an anti-TCRβV antibody molecule as described herein, e.g., a second anti-TCRβV antibody molecule;
    • (iii) inhibits, e.g., competitively inhibits, the binding of an anti-TCRβV antibody molecule as described herein, e.g., a second anti-TCRβV antibody molecule;
    • (vi) binds the same or an overlapping epitope with an anti-TCRβV antibody molecule as described herein, e.g., a second anti-TCRβV antibody molecule; or
    • (v) competes for binding, and/or binds the same epitope, with an anti-TCRβV antibody molecule as described herein, e.g., a second anti-TCRβV antibody molecule, wherein the second anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain complementarity determining region 1 (HC CDR1), a heavy chain complementarity determining region 2 (HC CDR2) and/or a heavy chain complementarity determining region 3 (HC CDR3) of SEQ ID NO: 1 or SEQ ID NO: 9; and/or a light chain complementarity determining region 1 (LC CDR1), a light chain complementarity determining region 2 (LC CDR2), and/or a light chain complementarity determining region 3 (LC CDR3) of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (a) a HC CDR1, a HC CDR2 and/or a HC CDR3 of SEQ ID NO: 15, SEQ ID NO: 23, SEQ ID NO: 24 or SEQ ID NO: 25; and/or (b) a LC CDR1, a LC CDR2, and/or a LC CDR3 of SEQ ID NO: 16, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO:30; or
    • (a) a LC CDR1, a LC CDR2 and/or a LC CDR3 of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11; and/or (b) a HC CDR1, a HC CDR2 and/or a HC CDR2 of SEQ ID NO: 1 or SEQ ID NO: 9.

In some embodiments, the anti-TCRβV antibody molecule binds the same or different TCRβV subfamily members.

In some embodiments, the multispecific molecule as provided herein comprises an antibody molecule chosen from a bispecific antibody molecule, a bivalent antibody molecule, or a biparatopic antibody molecule.

In some embodiments, the multispecific molecule as provided herein comprises a bispecific antibody molecule that binds to two different TCRβV subfamily members.

In some embodiments, the anti-TCRβV antibody molecule binds:

    • one or more of a TCRβ V6 subfamily member and one or more of a TCRβ V10 subfamily member; one or more of a TCRβ V6 subfamily member and one or more of a TCRβ V5 subfamily member; one or more of a TCRβ V6 subfamily member and one or more of a TCRβ V12 subfamily member; one or more of a TCRβ V10 subfamily member and one or more of a TCRβ V5 subfamily member; one or more of a TCRβ V10 subfamily member and one or more of a TCRβ V12 subfamily member; or one or more of a TCRβ V5 subfamily member and one or more of a TCRβ V12 subfamily member.

In another aspect, provided herein is a multispecific molecule, e.g., a bispecific molecule, comprising the anti-TCRβV antibody molecule as provided herein.

In another aspect, provided herein is an antibody molecule which binds, e.g., specifically binds, to a T cell receptor beta variable chain (TCRβV) region, wherein the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (a) a light chain variable region (VL) comprising:
    • (i) one, two or all of (e.g., three) a light chain complementarity determining region 1 (LC CDR1), a light chain complementarity determining region 2 (LC CDR2), and a light chain complementarity determining region 3 (LC CDR3) of SEQ ID NO: 10 or SEQ ID NO: 11; and
    • (ii) a framework region (FR) having at least 95% identity with one, two, three, or all of (e.g., four) a non-murine germline framework 1 (FR1), a non-murine germline framework region 2 (FR2), a non-murine germline framework region 3 (FR3), and a non-murine germline framework region 4 (FR4); and/or
    • (b) a heavy chain variable region (VH) comprising:
    • (i) one, two or all of (e.g., three) a heavy chain complementarity determining region 1 (HC CDR1), a heavy chain complementarity determining region 2 (HC CDR2) and a heavy chain complementarity determining region 3 (HC CDR3) of SEQ ID NO: 9; and
    • (ii) a framework region (FR) having at least 95% identity with one, two, three, or all of (e.g., four) a non-murine germline framework 1 (FR1), a non-murine germline framework region 2 (FR2), a non-murine germline framework region 3 (FR3), and a non-murine germline framework region 4 (FR4).

In some embodiments, the VL comprises an amino acid sequence having a consensus sequence of SEQ ID NO: 230.

In some embodiments, the VH comprises an amino acid sequence having a consensus sequence of SEQ ID NO: 231.

In some embodiments, the anti-TCRβV antibody molecule as provided herein binds to TCRβ V6, e.g., one or more of TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01, or a variant thereof.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of SEQ ID NO: 1 or SEQ ID NO: 9, or an amino acid sequence listed in Table 1; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11, or an amino acid sequence listed in Table 1.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11, or an amino acid sequence listed in Table 1.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of SEQ ID NO:1 or SEQ ID NO: 9, or an amino acid sequence listed in Table 1.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a VL comprising: a LC CDR1 amino acid sequence of SEQ ID NO: 6 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), a LC CDR2 amino acid sequence of SEQ ID NO:7 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), and/or a LC CDR3 amino acid sequence of SEQ ID NO:8 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof); and/or
    • (ii) a VH comprising: a HC CDR1 amino acid sequence of SEQ ID NO: 3 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), a HC CDR2 amino acid sequence of SEQ ID NO:4 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), and/or a HC CDR3 amino acid sequence of SEQ ID NO:5 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof).

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • a variable heavy chain (VH) of an amino acid sequence listed in Table 1, e.g., SEQ ID NO: 9 or SEQ ID NO: 1312, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to an amino acid sequence listed in Table 1, e.g., SEQ ID NO: 9 or SEQ ID NO: 1312; and/or
    • a variable light chain (VL) of an amino acid sequence listed in Table 1, e.g., SEQ ID NO: 10 or SEQ ID NO: 11 or SEQ ID NO: 1314, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to an amino acid sequence listed in Table 1, e.g., SEQ ID NO: 10 or SEQ ID NO: 11 or SEQ ID NO: 1314.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) the VH amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 1312;
    • (ii) an amino acid sequence having at least about 85%, 90%, 95%, or 99% sequence identity to the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 1312;
    • (iii) the VL amino acid sequence of SEQ ID NO: 10 or SEQ ID NO: 1314; and/or
    • (iv) an amino acid sequence having at least about 85%, 90%, 95%, or 99% sequence identity to the amino acid sequence of SEQ ID NO: 10 or SEQ ID NO: 1314.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) the VH amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 1312;
    • (ii) an amino acid sequence having at least about 85%, 90%, 95%, or 99% sequence identity to the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 1312;
    • (iii) the VL amino acid sequence of SEQ ID NO: 11 or SEQ ID NO: 1314; and/or
    • (iv) an amino acid sequence having at least about 85%, 90%, 95%, or 99% sequence identity to the amino acid sequence of SEQ ID NO: 11 or SEQ ID NO: 1314.

In some embodiments, the anti-TCRβV antibody molecule comprises a heavy chain comprising a framework region, e.g., framework region 3 (FR3), comprising one or both of:

    • (i) a Threonine at position 73, e.g., a substitution at position 73 according to Kabat numbering, e.g., a Glutamic Acid to Threonine substitution; or
    • (ii) a Glycine a position 94, e.g., a substitution at position 94 according to Kabat numbering, e.g., a Arginine to Glycine substitution;
    • wherein the substitution is relative to a human germline heavy chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule comprises a light chain comprising a framework region, e.g., framework region 1 (FR1), comprising a Phenyalanine at position 10, e.g., a substitution at position 10 according to Kabat numbering, e.g., a Serine to Phenyalanine substitution, wherein the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule comprises a light chain comprising a framework region, e.g., framework region 2 (FR2), comprising one or both of:

    • (i) a Histidine at position 36, e.g., a substitution at position 36 according to Kabat numbering, e.g., a Tyrosine to Histidine substitution; or
    • (ii) an Alanine at position 46, e.g., a substitution at position 46 according to Kabat numbering, e.g., a Arginine to Alanine substitution;
    • wherein the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule comprises a light chain comprising a framework region, e.g., framework region 3 (FR3), comprising a Phenyalanine at position 87, e.g., a substitution at position 87 according to Kabat numbering, e.g., a Tyrosine to Phenyalanine substitution, wherein the substitution is relative to a human germline light chain framework region sequence.

In another aspect, provided herein is an antibody molecule which binds, e.g., specifically binds, to a T cell receptor beta variable chain (TCRβV) region, wherein the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (a) a light chain variable region (VL) comprising:
    • (i) one, two or all of (e.g., three) a light chain complementarity determining region 1 (LC CDR1), a light chain complementarity determining region 2 (LC CDR2), and a light chain complementarity determining region 3 (LC CDR3) of a humanized B-H light chain (LC) of Table 2; and
    • (ii) a framework region (FR) having at least 95% identity with one, two, three or all (e.g., four) of a framework region 1 (FR1), a framework region 2 (FR2), a framework region 3 (FR3), and a framework region 4 (FR4) of a humanized B-H LC of Table 2; and/or
    • (b) a heavy chain variable region (VH) comprising:
    • (i) one, two or all of (e.g., three) a heavy chain complementarity determining region 1 (HC CDR1), a heavy chain complementarity determining region 2 (HC CDR2) and a heavy chain complementarity determining region 3 (HC CDR3) of a humanized B-H heavy chain (HC) of Table 2; and
    • (ii) a framework region (FR) having at least 95% identity with one, two, three or all (e.g., four) of a framework region 1 (FR1), a framework region 2 (FR2), a framework region 3 (FR3), and a framework region 4 (FR4) of a humanized B-H HC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule as provided herein binds to TCRβ V12, e.g., TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01, or a variant thereof.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of Antibody B-H listed in Table 2; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of Antibody B-H listed in Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11, or an amino acid sequence listed in Table 1.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all (e.g., three) of a HC CDR1, a HC CDR2 and a HC CDR3 of a humanized Antibody B-H listed in Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all (e.g., three) of a LC CDR1, a LC CDR2 and a LC CDR3 of a humanized Antibody B-H listed in Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • a VH sequence of a humanized Antibody B-H listed in Table 2, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VH of a humanized Antibody B-H listed in Table 2; and/or
    • a VL sequence of a humanized Antibody B-H listed in Table 2, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VL of a humanized Antibody B-H listed in Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with one of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H LC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with any two of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H LC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with any three of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H LC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with all of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H LC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with one of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H HC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with any two of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H HC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with any three of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H HC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with all of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H HC of Table 2.

In some embodiments, binding of the anti-TCRβV antibody molecule to the TCRβV region results in a cytokine profile that differs from a cytokine profile of a T cell engager that binds to a receptor or molecule other than a TCRβV region (“a non-TCRβV-binding T cell engager”).

In some embodiments, the non-TCRβV-binding T cell engager comprises an antibody that binds to a CD3 molecule (e.g., CD3 epsilon (CD3e) molecule); or a TCR alpha (TCRα) molecule.

In some embodiments, the cytokine profile of the first moiety comprises, one, two, three, four, five, six, seven, or all of the following:

    • (i) increased level, e.g., expression level, and/or activity of IL-2;
    • (ii) reduced level, e.g., expression level, and/or activity of IL-1β;
    • (iii) reduced level, e.g., expression level, and/or activity of IL-6;
    • (iv) reduced level, e.g., expression level, and/or activity of TNFα;
    • (v) reduced level, e.g., expression level, and/or activity of IL-10;
    • (vi) a delay, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more hours delay, in increased level, e.g., expression level, and/or activity of IL-2;
    • (vii) a delay, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 hours delay, in increased level, e.g., expression level, and/or activity of IFNg; or
    • (viii) increased level, e.g., expression level, and/or activity of IL-15, e.g., wherein (i)-(vii) are relative to the cytokine profile of the non-TCRβV-binding T cell engager.

In some embodiments, binding of the anti-TCRβV antibody molecule to the TCRβV region results in reduced cytokine storm, e.g., reduced cytokine release syndrome (CRS), as measured by an assay of Example 3, e.g., relative to the cytokine storm induced by the non-TCRβV-binding T cell engager.

In some embodiments, binding of the anti-TCRβV antibody molecule to the TCRβV region results in one, two, three or all of:

    • (ix) reduced T cell proliferation kinetics;
    • (x) cell killing, e.g., target cell killing, e.g. cancer cell killing, e.g., as measured by an assay of Example 4;
    • (xi) increased Natural Killer (NK) cell proliferation, e.g., expansion; or
    • (xii) expansion, e.g., at least about 1.1-10 fold expansion (e.g., at least about 1.1, 1.2, 1.3, 1.4, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, or 10 fold expansion), of a population of T cells having a memory-like phenotype,
    • e.g., relative to the non-TCRβV-binding T cell engager.

In some embodiments, the population of T cells having a memory-like phenotype comprises CD45RA+ CCR7− T cells, e.g., CD4+ and/or CD8+ T cells.

In some embodiments, binding of the anti-TCRβV antibody molecule to a TCRβV region results in a reduction of at least 2, 5, 10, 20, 50, 100, or 200 fold, or at least 2-200 fold (e.g., 5-150, 10-100, 20-50 fold) in the expression level and or activity of IL-10 as measured by an assay of Example 3.

In some embodiments, binding of the anti-TCRβV antibody molecule to a TCRβV region results in a reduction of at least 2, 5, 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 fold, or at least 2-1000 fold (e.g., 5-900, 10-800, 20-700, 50-600, 100-500, or 200-400 fold) in the expression level and or activity of IL-6 as measured by an assay of Example 3.

In some embodiments, binding of the anti-TCRβV antibody molecule to a TCRβV region results in a reduction of at least 2, 5, 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, or 2000 fold, or at least 2-2000 fold (e.g., 5-1000, 10-900, 20-800, 50-700, 100-600, 200-500, or 300-400 fold) in the expression level and or activity of TNFα as measured by an assay of Example 3.

In some embodiments, binding of the anti-TCRβV antibody molecule to a TCRβV region results in an increase of at least 2, 5, 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, or 2000 fold, or at least 2-2000 fold (e.g., 5-1000, 10-900, 20-800, 50-700, 100-600, 200-500, or 300-400 fold) in the expression level and or activity of IL-2 as measured by an assay of Example 3.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a single chain Fv (scFv) or a Fab.

In some embodiments, the anti-TCRβV antibody molecule binds to a conformational or a linear epitope on the T cell.

In some embodiments, the anti-TCRβV antibody molecule is a full antibody (e.g., an antibody that includes at least one, and preferably two, complete heavy chains, and at least one, and preferably two, complete light chains), or an antigen-binding fragment (e.g., a Fab, F(ab′)2, Fv, a single chain Fv fragment, a single domain antibody, a diabody (dAb), a bivalent antibody, or bispecific antibody or fragment thereof, a single domain variant thereof, or a camelid antibody).

In some embodiments, the anti-TCRβV antibody molecule comprises one or more heavy chain constant regions chosen from IgG1, IgG2, IgG3, IgGA1, IgGA2, IgG4, IgJ, IgM, IgD, or IgE, or a fragment thereof, e.g., as described in Table 3.

In some embodiments, the anti-TCRβV antibody molecule comprises a heavy chain constant region of an IgM or a fragment thereof, optionally wherein the IgM heavy chain constant region comprises the sequence of SEQ ID NO: 73, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments, the anti-TCRβV antibody molecule comprises a heavy chain constant region of an IgJ or a fragment thereof, optionally wherein the IgJ heavy chain constant region comprises the sequence of SEQ ID NO: 76 or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments, the anti-TCRβV antibody molecule comprises a heavy chain constant region of an IgGA1, or a fragment thereof, optionally wherein the IgGA1 heavy chain constant region comprises the sequence of SEQ ID NO: 74, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments, the anti-TCRβV antibody molecule comprises a heavy chain constant region of an IgGA2, or a fragment thereof, optionally wherein the IgGA2 heavy chain constant region comprises a sequence listed in Table 3, e.g., SEQ ID NO: 75, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments, the anti-TCRβV antibody molecule comprises a light chain constant region chosen from the light chain constant regions of kappa or lambda, or a fragment thereof, e.g., as described in Table 3.

In some embodiments, the anti-TCRβV antibody molecule comprises a light chain constant region of a kappa chain, or a fragment thereof, optionally wherein the kappa chain constant region comprises the sequence of SEQ ID NO: 39, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • (i) one or more heavy chain constant regions comprising a heavy chain constant region chosen from IgG1, IgG2, IgG3, IgGA1, IgGA2, IgG4, IgJ, IgM, IgD, or IgE, or a fragment thereof, e.g., as described in Table 3; and
    • (ii) a light chain constant region comprising a light chain constant region chosen from the light chain constant regions of kappa or lambda, or a fragment thereof, e.g., as described in Table 3.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • (i) a heavy chain constant region comprising:
    • (a) an IgM heavy chain constant region or a fragment thereof, comprising the sequence of SEQ ID NO: 73, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto;
    • (b) an IgGA1 heavy chain constant region or a fragment thereof, comprising the sequence of SEQ ID NO: 74, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto; or
    • (c) an IgGA2 heavy chain constant region or a fragment thereof, comprising the sequence of SEQ ID NO: 75, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto; and
    • (ii) a light chain constant region comprising a kappa chain constant region comprising the sequence of SEQ ID NO: 39, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto, optionally wherein, the anti-TCRβV antibody molecule further comprises an IgJ heavy chain constant region or a fragment thereof, wherein the IgJ heavy chain constant region comprises the sequence of SEQ ID NO: 76 or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments, the second moiety is a tumor-targeting moiety.

In some embodiments, the second moiety is a cytokine molecule.

In some embodiments, the second moiety is a stromal modifying moiety.

In some embodiments, the second moiety is an anti-TCRβV antibody molecule other than the first moiety.

In some embodiments, the first and/or second moiety binds to and activates an immune cell, e.g., an effector cell.

In some embodiments, the first and/or second moiety binds to, but does not activate an immune cell, e.g., an effector cell.

In some embodiments, the second moiety is chosen from an NK cell engager, a T cell engager other than an anti-TCRβV antibody molecule, a B cell engager, a dendritic cell engager, or a macrophage cell engager, or a combination thereof.

In some embodiments, the tumor-targeting moiety comprises an antibody molecule (e.g., Fab or scFv), a receptor molecule (e.g., a receptor, a receptor fragment or functional variant thereof), or a ligand molecule (e.g., a ligand, a ligand fragment or functional variant thereof), or a combination thereof, that binds to a cancer antigen.

In some embodiments, the tumor-targeting moiety binds to a cancer antigen present on a cancer, e.g., a hematological cancer, a solid tumor, a metastatic cancer, soft tissue tumor, metastatic lesion, or a combination thereof.

In some embodiments, the cancer antigen is a tumor antigen or stromal antigen, or a hematological antigen.

In some embodiments, the cancer antigen is chosen from: BCMA, CD19, CD20, CD22, FcRH5, PDL1, CD47, gangloside 2 (GD2), prostate stem cell antigen (PSCA), prostate specific membrane antigen (PMSA), prostate-specific antigen (PSA), carcinoembryonic antigen (CEA), Ron Kinase, c-Met, Immature laminin receptor, TAG-72, BING-4, Calcium-activated chloride channel 2, Cyclin-B1, 9D7, Ep-CAM, EphA3, Her2/neu, Telomerase, SAP-1, Survivin, NY-ESO-1/LAGE-1, PRAME, SSX-2, Melan-A/MART-1, Gp100/pmel17, Tyrosinase, TRP-1/-2, MC1R, β-catenin, BRCA1/2, CDK4, CML66, Fibronectin, p53, Ras, TGF-B receptor, AFP, ETA, MAGE, MUC-1, CA-125, BAGE, GAGE, NY-ESO-1, β-catenin, CDK4, CDC27, α actinin-4, TRP1/gp75, TRP2, gp100, Melan-A/MART1, gangliosides, WT1, EphA3, Epidermal growth factor receptor (EGFR), MART-2, MART-1, MUC1, MUC2, MUM1, MUM2, MUM3, NA88-1, NPM, OA1, OGT, RCC, RUI1, RUI2, SAGE, TRG, TRP1, TSTA, Folate receptor alpha, L1-CAM, CAIX, gpA33, GD3, GM2, VEGFR, Integrins (Integrin alphaVbeta3, Integrin alpha5Beta1), Carbohydrates (Le), IGF1R, EPHA3, TRAILR1, TRAILR2, RANKL, (FAP), TGF-beta, hyaluronic acid, collagen, e.g., collagen IV, tenascin C, or tenascin W.

In some embodiments, the tumor-targeting moiety is a BCMA targeting moiety or a FcRH5 targeting moiety.

In some embodiments, the cancer is a solid tumor including but not limited to: pancreatic (e.g., pancreatic adenocarcinoma) cancer, breast cancer, colorectal cancer, lung cancer (e.g., small or non-small cell lung cancer), skin cancer, ovarian cancer, or liver cancer.

In some embodiments, the cancer is a hematological cancer including, but not limited to: a B-cell or T cell malignancy, e.g., Hodgkin's lymphoma, Non-Hodgkin's lymphoma (e.g., B cell lymphoma, diffuse large B cell lymphoma, follicular lymphoma, chronic lymphocytic leukemia, mantle cell lymphoma, marginal zone B-cell lymphoma, Burkitt lymphoma, lymphoplasmacytic lymphoma, hairy cell leukemia), acute myeloid leukemia (AML), chronic myeloid leukemia, myelodysplastic syndrome, multiple myeloma, and acute lymphocytic leukemia.

In some embodiments, the cytokine molecule is chosen from interleukin-2 (IL-2), interleukin-7 (IL-7), interleukin-12 (IL-12), interleukin-15 (IL-15), interleukin-18 (IL-18), interleukin-21 (IL-21), or interferon gamma, or a fragment, variant or combination thereof.

In some embodiments, the cytokine molecule is a monomer or a dimer.

In some embodiments, the cytokine molecule further comprises a receptor dimerizing domain, e.g., an IL15Ralpha dimerizing domain.

In some embodiments, the cytokine molecule (e.g., IL-15) and the receptor dimerizing domain (e.g., an IL15Ralpha dimerizing domain) are not covalently linked, e.g., are non-covalently associated.

In some embodiments, the multispecific molecule as provided herein further comprises an immunoglobulin constant region (e.g., Fc region) chosen from the heavy chain constant regions of IgG1, IgG2, IgG3, IgGA1, IgGA2, IgG4, IgJ, IgM, IgD, or IgE, or a fragment thereof, optionally wherein, the heavy chain constant region comprises the heavy chain constant region of human IgG1, IgG2 or IgG4.

In some embodiments, the immunoglobulin constant region (e.g., an Fc region) is linked, e.g., covalently linked to, one or more of tumor-targeting moiety, the cytokine molecule, or the stromal modifying moiety.

In some embodiments, an interface of a first and second immunoglobulin chain constant regions (e.g., Fc region) is altered, e.g., mutated, to increase or decrease dimerization, e.g., relative to a non-engineered interface.

In some embodiments, the dimerization of the immunoglobulin chain constant region (e.g., Fc region) is enhanced by providing an Fc interface of a first and a second Fc region with one or more of: a paired cavity-protuberance (“knob-in-a hole”), an electrostatic interaction, or a strand-exchange, such that a greater ratio of heteromultimer:homomultimer forms, e.g., relative to a non-engineered interface.

In some embodiments, the multispecific molecule as provided herein further comprises a linker, e.g., a linker described herein, optionally wherein the linker is selected from: a cleavable linker, a non-cleavable linker, a peptide linker, a flexible linker, a rigid linker, a helical linker, or a non-helical linker.

In another aspect, provided herein is an isolated nucleic acid molecule comprising a nucleotide sequence encoding the anti-TCRβV antibody molecule as provided herein, or a nucleotide sequence having at least 75%, 80%, 85%, 90%, 95%, or 99% identity thereto.

In another aspect, provided herein is an isolated nucleic acid molecule comprising a nucleotide sequence encoding the multispecific molecule as provided herein, or a nucleotide sequence having at least 75%, 80%, 85%, 90%, 95%, or 99% identity thereto.

In another aspect, provided herein is a vector, e.g., an expression vector, comprising one or more of the nucleic acid molecules as provided herein.

In another aspect, provided herein is a cell, e.g., host cell, comprising the nucleic acid molecule as provided herein, or the vector as provided herein.

In another aspect, provided herein is a method of making, e.g., producing or manufacturing, the anti-TCRβV antibody molecule as provided herein, or the multispecific molecule as provided herein, comprising culturing the host cell as provided herein, under suitable conditions, e.g., conditions suitable expression of the anti-TCRβV antibody molecule or the multispecific molecule.

In another aspect, provided herein is a pharmaceutical composition comprising the anti-TCRβV antibody molecule as provided herein, or the multispecific molecule as provided herein, and a pharmaceutically acceptable carrier, excipient, or stabilizer.

In another aspect, provided herein is a method of modulating, e.g., enhancing, an immune response in a subject comprising administering to the subject an effective amount of an antibody molecule which binds (e.g., specifically binds) to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”).

In another aspect, provided herein is a method of modulating, e.g., enhancing, an immune response in a subject comprising administering to the subject an effective amount of the multispecific molecule as provided herein.

In some embodiments, the method comprises expanding, e.g., increasing the number of, an immune cell population in the subject.

In another aspect, provided herein is a method of expanding, e.g., increasing the number of, an immune cell population comprising, contacting the immune cell population with an effective amount of an antibody molecule which binds (e.g., specifically binds) to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”).

In another aspect, provided herein is a method of expanding, e.g., increasing the number of, an immune cell population comprising, contacting the immune cell population with an effective amount of the multispecific molecule as provided herein.

In some embodiments, the expansion occurs in vivo or ex vivo (e.g., in vitro).

In some embodiments, the immune cell population comprises a TCRβV expressing cell, e.g., a TCRβV+ cell.

In some embodiments, the TCRβV expressing cell is a T cell, e.g., a CD8+ T cell, a CD3+ T cell or a CD4+ T cell.

In some embodiments, the immune cell population comprises a T cell (e.g., a CD4 T cell, a CD8 T cell (e.g., an effector T cell, a T cell having a memory-like phenotype or a memory T cell (e.g., a memory effector T cell (e.g., TEM cell, e.g., TEMRA cell), or a tumor infiltrating lymphocyte (TIL).

In some embodiments, the immune cell population comprises a T cell, a Natural Killer cell, a B cell, or a myeloid cell.

In some embodiments, the immune cell population is obtained from a healthy subject.

In some embodiments, the immune cell population is obtained from a subject (e.g., from an apheresis sample from the subject) having a disease, e.g., a cancer, e.g., as described herein, optionally wherein the immune cell population comprises a tumor infiltrating lymphocyte (TIL).

In some embodiments, the method results in an expansion of at least 1.1-10 fold (e.g., at least 1.1, 1.2, 1.3, 1.4, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, or 10 fold expansion).

In some embodiments, the method as provided herein further comprises contacting the population of cells with an agent that promotes, e.g., increases, immune cell expansion.

In some embodiments, the method as provided herein further comprises contacting the population of cells with an immune checkpoint inhibitor, e.g., a PD-1 inhibitor.

In some embodiments, the method as provided herein further comprises contacting the population of cells with a 4-1BB (CD127) agonist, e.g., an anti-4-1BB antibody.

In some embodiments, the method as provided herein further comprises contacting the population of cells with a non-dividing population of cells, e.g., feeder cells, e.g., irradiated allogenic human PBMCs.

In some embodiments, the population of cells is expanded in an appropriate media (e.g., media described herein) that includes one or more cytokines, e.g., IL-2, IL-7, IL-15, or a combination thereof.

In some embodiments, the population of cells is expanded for a period of at least about 4 hours, 6 hours, 10 hours, 12 hours, 15 hours, 18 hours, 20 hours, or 22 hours, or for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 1,6 17, 18, 19, 20 or 21 days, or for at least about 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks or 8 weeks.

In some embodiments, expansion of the population of immune cells, is compared to expansion of a similar population of cells with an antibody that binds to: a CD3 molecule, e.g., CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments, expansion of the population of immune cells, is compared to expansion of a similar population of cells not contacted with the anti-TCRβV antibody molecule, or multi specific molecule comprising the anti-TCRβV antibody molecule.

In some embodiments, expansion of the population of T cells having a memory-like phenotype, e.g., CD45RA+ CCR7− cells (e.g., memory effector T cells, e.g., TEM cells, e.g., TEMRA cells), is compared to expansion of a similar population of cells with an antibody that binds to: a CD3 molecule, e.g., CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments, the population of expanded T cells having a memory-like phenotype, e.g., effector memory cells, comprises cells which:

    • (i) have a detectable level of CD45RA, e.g., express or re-express CD45RA;
    • (ii) have low or no expression of CCR7; and/or
    • (iii) have a detectable level of CD95, e.g., express CD95,
    • e.g., a population of CD45RA+, CCR7−, CD95+ T cells, optionally wherein the T cells comprise CD3+, CD4+ or CD8+ T cells.

In some embodiments, the method results in expansion of, e.g., selective or preferential expansion of, T cells expressing a T cell receptor (TCR) comprising a TCR alpha and/or TCR beta molecule, e.g., TCR alpha-beta T cells (αβ T cells).

In some embodiments, the method results in expansion of αβT cells over expansion of T cells expressing a TCR comprising a TCR gamma and/or TCR delta molecule, e.g., TCR gamma-delta T cells (γδ T cells).

In another aspect, provided herein is a method of treating a disease, e.g., cancer, in a subject comprising administering to the subject an effective amount of an antibody molecule which binds (e.g., specifically binds) to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”), thereby treating the cancer.

In another aspect, provided herein is a composition comprising an antibody molecule which binds (e.g., specifically binds) to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”), for use in treating a disease, e.g., cancer, in a subject.

In another aspect, provided herein is a composition comprising an antibody molecule which binds (e.g., specifically binds) to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”), for use in the manufacture of a medicament for treating a disease, e.g., cancer, in a subject.

In another aspect, provided herein is a method of treating a disease, e.g., cancer, in a subject comprising administering to the subject an effective amount of the multispecific molecule as provided herein, thereby treating the cancer.

In another aspect, provided herein is a composition comprising the multispecific molecule as provided herein, for use in treating a disease, e.g., cancer, in a subject.

In another aspect, provided herein is a composition comprising the multispecific molecule as provided herein, for use in the manufacture of a medicament for treating a disease, e.g., cancer, in a subject.

In another aspect, provided herein is a method of treating, e.g., preventing or reducing, cytokine release syndrome (CRS) and/or neurotoxicity (NT) in a subject, e.g., CRS and/or NT associated with a treatment, e.g., a previously administered treatment, comprising administering to the subject an effective amount of an antibody molecule which binds (e.g., specifically binds) to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”), thereby preventing CRS and/or NT in the subject.

In another aspect, provided herein is a method of treating, e.g., preventing or reducing, cytokine release syndrome (CRS) and/or neurotoxicity (NT) in a subject, e.g., CRS and/or NT associated with a treatment, e.g., a previously administered treatment, comprising administering to the subject an effective amount the multispecific molecule as provided herein, thereby preventing CRS and/or NT in the subject.

In another aspect, provided herein is a method of targeting a therapy, e.g., treatment, to a T cell in a subject having a disease, e.g., cancer, comprising administering an effective amount of:

    • (i) an antibody molecule which binds (e.g., specifically binds) to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”); and
    • (ii) the therapy, e.g., a tumor targeting therapy (e.g., an antibody that binds to a cancer antigen),
    • e.g., as described herein,
    • thereby targeting the therapy to the T cell in the subject.

In another aspect, provided herein is a method of targeting a therapy, e.g., treatment, to a T cell in a subject having a disease, e.g., cancer, comprising administering an effective amount of:

    • (i) the multispecific molecule as provided herein; and
    • (ii) the therapy, e.g., a tumor targeting therapy (e.g., an antibody that binds to a cancer antigen),
    • e.g., as described herein,
    • thereby targeting the therapy to the T cell in the subject.

In some embodiments, the method results in: reduced cytokine release syndrome (CRS) (e.g., lesser duration of CRS or no CRS), or a reduced severity of CRS (e.g., absence of severe CRS, e.g., CRS grade 4 or 5) compared to administration of (ii) alone.

In some embodiments, the anti-TCRβV antibody or the multispecific molecule is administered concurrently with or after the administration of the treatment associated with CRS.

In another aspect, provided herein is a method of treating a subject having a cancer, the method comprising:

    • acquiring a value of the status of a TCRβV subfamily for the subject, wherein said value comprises a measure of the presence of, e.g., level or activity of, a TCRβV molecule in a sample from the subject, and
    • administering an effective amount of an antibody molecule which binds (e.g., specifically binds) to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”); to the subject, thereby treating the subject.

In another aspect, provided herein is a method of treating a subject having a cancer, the method comprising:

    • acquiring a value of the status of a TCRβV subfamily for the subject, wherein said value comprises a measure of the presence of, e.g., level or activity of, a TCRβV molecule in a sample from the subject, and
    • administering an effective amount of the multispecific molecule as provided herein to the subject, thereby treating the subject.

In another aspect, provided herein is a method of treating a subject having a cancer, the method comprising administering an effective amount of an antibody molecule which binds (e.g., specifically binds) to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”) to the subject, wherein the subject has a higher, e.g., increased, level or activity of one or more TCRβV subfamilies, e.g., as described herein, compared to a reference level or activity of one or more TCRβV subfamilies, e.g., in a healthy subject, e.g., a subject not having a cancer.

In another aspect, provided herein is a method of treating a subject having a cancer, the method comprising administering an effective amount of the multispecific molecule as provided herein to the subject, wherein the subject has a higher, e.g., increased, level or activity of one or more TCRβV subfamilies, e.g., as described herein, compared to a reference level or activity of one or more TCRβV subfamilies, e.g., in a healthy subject, e.g., a subject not having a cancer.

In another aspect, provided herein is a method of expanding a population of immune effector cells from a subject having a cancer, the method comprising:

    • (i) isolating a biological sample comprising a population of immune effector cells from the subject; e.g., a peripheral blood sample, biopsy sample, or bone marrow sample;
    • (ii) acquiring a value of the status of one or more TCRβV subfamilies for the subject, e.g., in the biological sample from the subject, wherein said value comprises a measure of the presence of, e.g., level or activity of, a TCRβV subfamily in a sample from the subject compared to a reference value, e.g., a sample from a health subject, wherein a value that is higher, e.g., increased, in the subject relative to the reference, e.g., healthy subject, is indicative of the presence of cancer in the subject, and
    • (iii) contacting the biological sample comprising a population of immune effector cells with an anti-TCRβV antibody molecule, e.g., as described herein.

In some embodiments, the method as provided herein further comprises administering the population of immune effector cells contacted with the anti-TCRβV antibody molecule to the subject.

In another aspect, provided herein is a method of expanding a population of immune effector cells from a subject having a cancer, the method comprising:

    • (i) isolating a biological sample comprising a population of immune effector cells from the subject; e.g., a peripheral blood sample, biopsy sample, or bone marrow sample;
    • (ii) acquiring a value of the status of one or more TCRβV subfamilies for the subject, e.g., in the biological sample from the subject, wherein said value comprises a measure of the presence of, e.g., level or activity of, a TCRβV subfamily in a sample from the subject compared to a reference value, e.g., a sample from a health subject, wherein a value that is higher, e.g., increased, in the subject relative to the reference, e.g., healthy subject, is indicative of the presence of cancer in the subject, and
    • (iii) contacting the biological sample comprising a population of immune effector cells with the multispecific molecule as provided herein.

In some embodiments, the method as provided herein further comprises administering the population of immune effector cells contacted with the multispecific molecule to the subject.

In some embodiments, the method as provided herein comprises measuring T cell function (e.g., cytotoxic activity, cytokine secretion, or degranulation) in the population of immune effector cells, e.g., compared to a reference population, e.g., an otherwise similar population not contacted with the anti-TCRβV antibody molecule or a population of immune effector cells obtained from a healthy subject (e.g., a subject that does not have a cancer).

In some embodiments, the biological sample comprising the population of immune effector cells is contacted with an anti-TCRβV antibody molecule or a multispecific molecule that binds to the one or more TCRβV subfamilies (e.g., the same TCRβV subfamily) identified as being higher, e.g., increased, in the biological sample.

In some embodiments, the biological sample comprising the population of immune effector cells is contacted with an anti-TCRβV antibody molecule or a multispecific molecule that does not bind to the one or more TCRβV subfamilies (e.g., a different TCRβV subfamily) identified as being higher, e.g., increased, in the biological sample.

In some embodiments, the cancer is a solid tumor including but not limited to: melanoma, pancreatic (e.g., pancreatic adenocarcinoma) cancer, breast cancer, colorectal cancer (CRC), lung cancer (e.g., small or non-small cell lung cancer), skin cancer, ovarian cancer, or liver cancer.

In some embodiments, the cancer is a hematological cancer including, but not limited to: a B-cell or T cell malignancy, e.g., Hodgkin's lymphoma, Non-Hodgkin's lymphoma (e.g., B cell lymphoma, diffuse large B cell lymphoma (DLBCL), follicular lymphoma, chronic lymphocytic leukemia (B-CLL), mantle cell lymphoma, marginal zone B-cell lymphoma, Burkitt lymphoma, lymphoplasmacytic lymphoma, hairy cell leukemia), acute myeloid leukemia (AML), chronic myeloid leukemia, myelodysplastic syndrome, multiple myeloma, and acute lymphocytic leukemia.

In some embodiments, the cancer is B-CLL and the TCRβV molecule comprises:

    • (i) TCRβ V6 subfamily comprising, e.g., TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01;
    • (ii) TCRβ V5 subfamily comprising TCRβ V5-6*01, TCRβ V5-4*01, or TCRβ V5-8*01;
    • (iii) TCRβ V3 subfamily comprising TCRβ V3-1*01;
    • (iv) TCRβ V2 subfamily comprising TCRβ V2*01; or
    • (v) TCRβ V19 subfamily comprising TCRβ V19*01, or TCRβ V19*02.

In some embodiments, the cancer is melanoma and the TCRβV molecule comprises the TCRβ V6 subfamily comprising, e.g., TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01.

In some embodiments, the cancer is DLBCL and the TCRβV molecule comprises:

    • (i) TCRβ V13 subfamily comprising TCRβ V13*01;
    • (ii) TCRβ V3 subfamily comprising TCRβ V3-1*01; or
    • (iii) TCRβ V23 subfamily.

In some embodiments, the cancer is CRC and the TCRβV molecule comprises:

    • (i) TCRβ V19 subfamily comprising TCRβ V19*01, or TCRβ V19*02
    • (ii) TCRβ V12 subfamily comprising TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01
    • (iii) TCRβ V16 subfamily comprising TCRβ V16*01; or
    • (iv) TCRβ V21 subfamily.

In some embodiments, the tumor comprises an antigen, e.g., a tumor antigen, e.g., a tumor associated antigen or a neoantigen; and/or the one or more TCRβV subfamilies recognize, e.g., bind to, the tumor antigen.

In some embodiments, the sample comprises a blood sample, e.g., a peripheral blood sample, a biopsy, e.g., a tumor biopsy, or a bone marrow sample.

In some embodiments, the sample comprises a biological sample comprising immune cells, e.g., TCRBV expressing cells (e.g., TCRBV+ cells), T cells, or NK cells.

In some embodiments, the T cells comprise a CD4 T cell, a CD8 T cell, (e.g., an effector T cell or a memory T cell (e.g., a memory effector T cell (e.g., TEM cell, e.g., TEMRA cell), or a tumor infiltrating lymphocyte (TIL).

In some embodiments, the method results in an expansion, e.g., in vivo or ex vivo expansion, of at least 1.1-1000 fold, e.g., 1.1-10, 10-100, 100-200, 200-300, 300-400, 400-500, 500-600, 600-700, 700-800, 800-900, or 900-1000 fold expansion of an immune effector cell population comprising a TCRVB expressing immune effector cell, e.g., T cell.

In some embodiments, the population of cells is expanded in an appropriate media (e.g., media described herein) that includes one or more cytokines, e.g., IL-2, IL-7, IL-15, or a combination thereof.

In some embodiments, the population of cells is expanded for a period of at least about 4 hours, 6 hours, 10 hours, 12 hours, 15 hours, 18 hours, 20 hours, or 22 hours, or for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 1,6 17, 18, 19, 20 or 21 days, or for at least about 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks or 8 weeks.

In some embodiments, expansion of the population of immune cells, is compared to expansion of a similar population of cells with an antibody that binds to: a CD3 molecule, e.g., CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments, expansion of the population of immune cells, is compared to expansion of a similar population of cells not contacted with the anti-TCRβV antibody molecule.

In some embodiments, expansion of the population of T cells having a memory-like phenotype, e.g., memory effector T cells, e.g., TEM cells, e.g., TEMRA cells, is compared to expansion of a similar population of cells with an antibody that binds to: a CD3 molecule, e.g., CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments, the population of expanded T cells having a memory-like phenotype, e.g., effector memory cells, comprises cells which:

    • (i) have a detectable level of CD45RA, e.g., express or re-express CD45RA;
    • (ii) have low or no expression of CCR7; and/or
    • (iii) have a detectable level of CD95, e.g., express CD95,
    • e.g., a population of CD45RA+, CCR7−, CD95+ T cells, optionally wherein the T cells comprise CD3+, CD4+ or CD8+ T cells.

In some embodiments, the method results in expansion of, e.g., selective or preferential expansion of, T cells expressing a T cell receptor (TCR) comprising a TCR alpha and/or TCR beta molecule, e.g., TCR alpha-beta T cells (αβ T cells).

In some embodiments, the method results in expansion of αβT cells over expansion of T cells expressing a TCR comprising a TCR gamma and/or TCR delta molecule, e.g., TCR gamma-delta T cells (γδ T cells).

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all of a LC CDR1, a LC CDR2 and a LC CDR3 of a VL disclosed in Tables 1, 2, 10, 11, 12 or 13, e.g., SEQ ID NO: 1314, SEQ ID NO: 2, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 16, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO:30

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all of a HC CDR1, a HC CDR2 and a HC CDR3 of a VH disclosed in Tables 1, 2, 10, 11, 12 or 13, e.g., SEQ ID NO: 1312, SEQ ID NO:1, SEQ ID NO: 9, SEQ ID NO: 15, SEQ ID NO: 23, SEQ ID NO: 24 or SEQ ID NO: 25.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a VL comprising: a LC CDR1 amino acid sequence of SEQ ID NO: 20 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), a LC CDR2 amino acid sequence of SEQ ID NO:21 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), and/or a LC CDR3 amino acid sequence of SEQ ID NO:22 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof); and/or
    • (ii) a VH comprising: a HC CDR1 amino acid sequence of SEQ ID NO: 17 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), a HC CDR2 amino acid sequence of SEQ ID NO:18 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), and/or a HC CDR3 amino acid sequence of SEQ ID NO:19 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof).

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • a variable heavy chain (VH) of SEQ ID NO: 23, SEQ ID NO: 24 or SEQ ID NO: 25, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity thereto; and/or
    • a variable light chain (VL) of SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO:30, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising

    • (i) a VL comprising: a LC CDR1 amino acid sequence of SEQ ID NO: 6 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), a LC CDR2 amino acid sequence of SEQ ID NO:7 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), and/or a LC CDR3 amino acid sequence of SEQ ID NO: 8 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof); and/or
    • (ii) a VH comprising: a HC CDR1 amino acid sequence of SEQ ID NO: 3 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), a HC CDR2 amino acid sequence of SEQ ID NO:4 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof), and/or a HC CDR3 amino acid sequence of SEQ ID NO:5 (or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, e.g., substitutions, additions or deletions thereof).

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • a variable heavy chain (VH) of SEQ ID NO: 1 or SEQ ID NO: 9 or SEQ ID NO: 1312, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity thereto; and/or
    • a variable light chain (VL) of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO:11 or SEQ ID NO: 1314, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments, the anti-TCRβV antibody molecule comprises a light chain comprising a framework region, e.g., framework region 1 (FR1), comprising one, two or all (e.g., three) of:

    • (i) an Aspartic Acid at position 1, e.g., a substitution at position 1 according to Kabat numbering, e.g., a Alanine to Aspartic Acid substitution; or
    • (ii) an Asparagine at position 2, e.g., a substitution at position 2 according to Kabat numbering, e.g., a Isoleucine to Asparagine, a Serine to Asparagine, or a Tyrosine to Asparagine substitution; or
    • (iii) a Leucine at position 4, e.g., a substitution at position 4 according to Kabat numbering, e.g., a Methionine to Leucine substitution,
    • wherein the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule comprises a light chain comprising a framework region, e.g., framework region 3 (FR3), comprising one, two or all (e.g., three) of:

    • (i) a Glycine at position 66, e.g., a substitution at position 66 according to Kabat numbering, e.g., a Lysine to Glycine, or a Serine to Glycine substitution; or
    • (ii) an Asparagine at position 69, e.g., a substitution at position 69 according to Kabat numbering, e.g., a Threonine to Asparagine substitution; or
    • (iii) a Tyrosine at position 71, e.g., a substitution at position 71 according to Kabat numbering, e.g., a Phenylalanine to Tyrosine, or Alanine to Tyrosine substitution,
    • wherein the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, binding of the anti-TCRβV antibody molecule to the TCRβV region results in a cytokine profile that differs from a cytokine profile of a T cell engager that binds to a receptor or molecule other than a TCRβV region (“a non-TCRβV-binding T cell engager”).

In some embodiments, the non-TCRβV-binding T cell engager comprises an antibody that binds to a CD3 molecule (e.g., CD3 epsilon (CD3e) molecule); or a TCR alpha (TCRα) molecule.

In some embodiments, the cytokine profile of the first moiety comprises, one, two, three, four, five, six, seven, or all of the following:

    • (i) increased level, e.g., expression level, and/or activity of IL-2;
    • (ii) reduced level, e.g., expression level, and/or activity of IL-1β;
    • (iii) reduced level, e.g., expression level, and/or activity of IL-6;
    • (iv) reduced level, e.g., expression level, and/or activity of TNFα;
    • (v) reduced level, e.g., expression level, and/or activity of IL-10;
    • (vi) a delay, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more hours delay, in increased level, e.g., expression level, and/or activity of IL-2;
    • (vii) a delay, e.g., at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 hours delay, in increased level, e.g., expression level, and/or activity of IFNg; or
    • (viii) increased level, e.g., expression level, and/or activity of IL-15, e.g., wherein (i)-(vii) are relative to the cytokine profile of the non-TCRβV-binding T cell engager.

In some embodiments, binding of the anti-TCRβV antibody molecule to the TCRβV region results in reduced cytokine storm, e.g., reduced cytokine release syndrome (CRS), as measured by an assay of Example 3, e.g., relative to the cytokine storm induced by the non-TCRβV-binding T cell engager.

In some embodiments, binding of the anti-TCRβV antibody molecule to the TCRβV region results in one, two, three or all of:

    • (ix) reduced T cell proliferation kinetics;
    • (x) cell killing, e.g., target cell killing, e.g. cancer cell killing, e.g., as measured by an assay of Example 4;
    • (xi) increased Natural Killer (NK) cell proliferation, e.g., expansion; or
    • (xii) expansion, e.g., at least about 1.1-10 fold expansion (e.g., at least about 1.1, 1.2, 1.3, 1.4, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, or 10 fold expansion), of a population of T cells having a memory-like phenotype,
    • e.g., relative to the non-TCRβV-binding T cell engager.

In some embodiments, the anti-TCRβV antibody molecule binds to an outward facing region (e.g., epitope) on a TCRβV protein, e.g., as depicted by the circled area in FIG. 24A.

In some embodiments, the outward facing region on the TCRβV protein comprises a structurally conserved region of TCRβV, e.g., a region of TCRβV having a similar structure across one or more TCRβV subfamilies.

In some embodiments, the method further comprises administering (e.g., sequentially, simultaneously or concurrently) a second agent, e.g., therapeutic agent, e.g., as described herein.

In some embodiments, the second agent, e.g., therapeutic agent, comprises a chemotherapeutic agent, a biologic agent, hormonal therapy), radiation, or surgery.

In some embodiments, the disease is a cancer, e.g., a solid tumor or a hematological cancer, or a metastatic lesion.

In some embodiments, the cancer antigen is BCMA or FcRH5.

In some aspects, provided herein is, inter alia, a multispecific molecule, optionally a bispecific molecule, comprising a first moiety, optionally a first immune cell engager, comprising an antibody molecule which binds, optionally specifically binds, to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”), wherein binding of the first moiety to the TCRβV region results in a cytokine profile that differs from a cytokine profile of a T cell engager that binds to a receptor or molecule other than a TCRβV region (“a non-TCRβV-binding T cell engager”).

In some embodiments, the multispecific molecule as provided herein comprises a second moiety which comprises one or more of: a tumor-targeting moiety, a cytokine molecule, a stromal modifying moiety, or an anti-TCRβV antibody molecule other than the first moiety.

In some embodiments, the first moiety comprising the anti-TCRβV antibody molecule comprises an Fc region comprising a variant, optionally an Fc variant described in Table 21, optionally an Asn297Ala (N297A) mutation or a Leu234A1a/Leu235Ala (LALA) mutation.

In some embodiments, the non-TCRβV-binding T cell engager comprises an antibody that binds to a CD3 molecule, optionally CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments, the cytokine profile of the first moiety comprises, one, two, three, four, five, six, seven, or all of the following:

    • (i) increased level, optionally expression level, and/or activity of IL-2;
    • (ii) reduced level, optionally expression level, and/or activity of IL-10;
    • (iii) reduced level, optionally expression level, and/or activity of IL-6;
    • (iv) reduced level, optionally expression level, and/or activity of TNFα;
    • (v) reduced level, optionally expression level, and/or activity of IL-10;
    • (vi) a delay, optionally at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more hours delay, in increased level, optionally expression level, and/or activity of IL-2;
    • (vii) a delay, optionally at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 hours delay, in increased level, optionally expression level, and/or activity of IFNg; or
    • (viii) increased level, optionally expression level, and/or activity of IL-15, optionally wherein (i)-(viii) are relative to the cytokine profile of the non-TCRβV-binding T cell engager.

In some embodiments, binding of the first moiety to the TCRβV region results in reduced cytokine storm, optionally reduced cytokine release syndrome (CRS), as measured by an assay of Example 3, optionally relative to the cytokine storm induced by the non-TCRβV-binding T cell engager.

In some embodiments, binding of the first moiety to the TCRβV region results in one, two, three or all of:

    • (ix) reduced T cell proliferation kinetics;
    • (x) cell killing, optionally target cell killing, optionally cancer cell killing, optionally as measured by an assay of Example 4;
    • (xi) increased Natural Killer (NK) cell proliferation, optionally expansion; or
    • (xii) expansion, optionally at least about 1.1-10 fold expansion, optionally at least about 1.1, 1.2, 1.3, 1.4, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, or 10 fold expansion, of a population of T cells having a memory-like phenotype,
    • optionally wherein (ix)-(xii) are relative to the non-TCRβV-binding T cell engager.

In some embodiments, the population of T cells having a memory-like phenotype comprises CD45RA+ CCR7− T cells, optionally CD4+ and/or CD8+ T cells.

In some embodiments, the first moiety binds to one or more of a TCRβV subfamily chosen from:

    • (i) TCRβ V6 subfamily comprising, optionally one or more of TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01;
    • (ii) TCRβ V10 subfamily comprising, optionally one or more of TCRβ V10-1*01, TCRβ V10-1*02, TCRβ V10-3*01 or TCRβ V10-2*01;
    • (iii) TCRβ V5 subfamily comprising, optionally one or more of TCRβ V5-6*01, TCRβ V5-4*01, TCRβ V5-1*01 or TCRβ V5-8*01;
    • (iv) TCRβ V12 subfamily comprising, optionally one or more of TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01;
    • (v) TCRβ V27 subfamily;
    • (vi) TCRβ V28 subfamily;
    • (vii) TCRβ V4 subfamily comprising, optionally one or more of TCRβ V4-1, TCRβ V4-2 or TCRβ V4-3;
    • (viii) TCRβ V19 subfamily;
    • (ix) TCRβ V9 subfamily; or
    • (x) TCRβ V11 subfamily comprising, optionally TCRβ V11-2.

In some embodiments, the anti-TCRβV antibody molecule:

    • (i) binds specifically to an epitope on TCRβV, optionally the same or similar epitope as the epitope recognized by an anti-TCRβV antibody molecule as described herein, optionally a second anti-TCRβV antibody molecule;
    • (ii) shows the same or similar binding affinity or specificity, or both, as an anti-TCRβV antibody molecule as described herein, optionally a second anti-TCRβV antibody molecule;
    • (iii) inhibits, optionally competitively inhibits, the binding of an anti-TCRβV antibody molecule as described herein, optionally a second anti-TCRβV antibody molecule;
    • (iv) binds the same or an overlapping epitope with an anti-TCRβV antibody molecule as described herein, optionally a second anti-TCRβV antibody molecule; or
    • (v) competes for binding, and/or binds the same epitope, with an anti-TCRβV antibody molecule as described herein, optionally a second anti-TCRβV antibody molecule,
    • wherein the second anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain complementarity determining region 1 (HC CDR1), a heavy chain complementarity determining region 2 (HC CDR2) and/or a heavy chain complementarity determining region 3 (HC CDR3) of SEQ ID NO: 1 or SEQ ID NO: 9; and/or a light chain complementarity determining region 1 (LC CDR1), a light chain complementarity determining region 2 (LC CDR2), and/or a light chain complementarity determining region 3 (LC CDR3) of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (a) a HC CDR1, a HC CDR2 and/or a HC CDR3 of SEQ ID NO: 15, SEQ ID NO: 23, SEQ ID NO: 24 or SEQ ID NO: 25; and/or (b) a LC CDR1, a LC CDR2, and/or a LC CDR3 of SEQ ID NO: 16, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO:30; or
    • (a) a LC CDR1, a LC CDR2 and/or a LC CDR3 of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11; and/or (b) a HC CDR1, a HC CDR2 and/or a HC CDR2 of SEQ ID NO: 1 or SEQ ID NO: 9.

In some embodiments, the anti-TCRβV antibody molecule binds the same or different TCRβV subfamily members.

In some embodiments, the multispecific molecule as provided herein comprises an antibody molecule chosen from a bispecific antibody molecule, a bivalent antibody molecule, or a biparatopic antibody molecule.

In some embodiments, the multispecific molecule as provided herein comprises a bispecific antibody molecule that binds to two different TCRβV subfamily members.

In some embodiments, the anti-TCRβV antibody molecule binds:

    • one or more of a TCRβ V6 subfamily member and one or more of a TCRβ V10 subfamily member;
    • one or more of a TCRβ V6 subfamily member and one or more of a TCRβ V5 subfamily member;
    • one or more of a TCRβ V6 subfamily member and one or more of a TCRβ V12 subfamily member;
    • one or more of a TCRβ V10 subfamily member and one or more of a TCRβ V5 subfamily member;
    • one or more of a TCRβ V10 subfamily member and one or more of a TCRβ V12 subfamily member; or
    • one or more of a TCRβ V5 subfamily member and one or more of a TCRβ V12 subfamily member.

In another aspect, provided herein is a multispecific molecule, optionally a bispecific molecule, comprising the anti-TCRβV antibody molecule as provided herein.

In another aspect, provided herein is an antibody molecule which binds, optionally specifically binds, to a T cell receptor beta variable chain (TCRβV) region, wherein the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (a) a light chain variable region (VL) comprising:
    • (i) one, two or all of, optionally three, a light chain complementarity determining region 1 (LC CDR1), a light chain complementarity determining region 2 (LC CDR2), and a light chain complementarity determining region 3 (LC CDR3) of SEQ ID NO: 10 or SEQ ID NO: 11; and
    • (ii) a framework region (FR) having at least 95% identity with one, two, three, or all of, optionally four, a non-murine germline framework 1 (FR1), a non-murine germline framework region 2 (FR2), a non-murine germline framework region 3 (FR3), and a non-murine germline framework region 4 (FR4); and/or
    • (b) a heavy chain variable region (VH) comprising:
    • (i) one, two or all of, optionally three, a heavy chain complementarity determining region 1 (HC CDR1), a heavy chain complementarity determining region 2 (HC CDR2) and a heavy chain complementarity determining region 3 (HC CDR3) of SEQ ID NO: 9; and
    • (ii) a framework region (FR) having at least 95% identity with one, two, three, or all of, optionally four, a non-murine germline framework 1 (FR1), a non-murine germline framework region 2 (FR2), a non-murine germline framework region 3 (FR3), and a non-murine germline framework region 4 (FR4).

In some embodiments, the VL comprises an amino acid sequence having a consensus sequence of SEQ ID NO: 230.

In some embodiments, the VH comprises an amino acid sequence having a consensus sequence of SEQ ID NO: 231.

In some embodiments, the anti-TCRβV antibody molecule as provided herein binds to TCRβ V6, optionally one or more of TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01, or a variant thereof.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of SEQ ID NO: 1 or SEQ ID NO: 9, or an amino acid sequence listed in Table 1; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11, or an amino acid sequence listed in Table 1.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all, optionally three, of a LC CDR1, a LC CDR2 and a LC CDR3 of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11, or an amino acid sequence listed in Table 1.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all, optionally three, of a HC CDR1, a HC CDR2 and a HC CDR3 of SEQ ID NO:1 or SEQ ID NO: 9, or an amino acid sequence listed in Table 1.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a VL comprising: a LC CDR1 amino acid sequence of SEQ ID NO: 6 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof, a LC CDR2 amino acid sequence of SEQ ID NO:7 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof, and/or a LC CDR3 amino acid sequence of SEQ ID NO:8 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof; and/or
    • (ii) a VH comprising: a HC CDR1 amino acid sequence of SEQ ID NO: 3 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof, a HC CDR2 amino acid sequence of SEQ ID NO:4 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof, and/or a HC CDR3 amino acid sequence of SEQ ID NO:5 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • a variable heavy chain (VH) of an amino acid sequence listed in Table 1, optionally SEQ ID NO: 9 or SEQ ID NO: 1312, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to an amino acid sequence listed in Table 1, optionally SEQ ID NO: 9 or SEQ ID NO: 1312; and/or
    • a variable light chain (VL) of an amino acid sequence listed in Table 1, optionally SEQ ID NO: 10 or SEQ ID NO: 11 or SEQ ID NO: 1314, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to an amino acid sequence listed in Table 1, optionally SEQ ID NO: 10 or SEQ ID NO: 11 or SEQ ID NO: 1314.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) the VH amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 1312;
    • (ii) an amino acid sequence having at least about 85%, 90%, 95%, or 99% sequence identity to the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 1312;
    • (iii) the VL amino acid sequence of SEQ ID NO: 10 or SEQ ID NO: 1314; and/or
    • (iv) an amino acid sequence having at least about 85%, 90%, 95%, or 99% sequence identity to the amino acid sequence of SEQ ID NO: 10 or SEQ ID NO: 1314.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) the VH amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 1312;
    • (ii) an amino acid sequence having at least about 85%, 90%, 95%, or 99% sequence identity to the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 1312;
    • (iii) the VL amino acid sequence of SEQ ID NO: 11 or SEQ ID NO: 1314; and/or
    • (iv) an amino acid sequence having at least about 85%, 90%, 95%, or 99% sequence identity to the amino acid sequence of SEQ ID NO: 11 or SEQ ID NO: 1314.

In some embodiments, the anti-TCRβV antibody molecule comprises a heavy chain comprising a framework region, optionally framework region 3 (FR3), comprising one or both of:

    • (i) a Threonine at position 73, optionally a substitution at position 73 according to Kabat numbering, optionally a Glutamic Acid to Threonine substitution; or
    • (ii) a Glycine a position 94, optionally a substitution at position 94 according to Kabat numbering, optionally a Arginine to Glycine substitution;
    • wherein the substitution is relative to a human germline heavy chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule comprises a light chain comprising a framework region, optionally framework region 1 (FR1), comprising a Phenyalanine at position 10, optionally a substitution at position 10 according to Kabat numbering, optionally a Serine to Phenyalanine substitution, wherein the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule comprises a light chain comprising a framework region, optionally framework region 2 (FR2), comprising one or both of:

    • (i) a Histidine at position 36, optionally a substitution at position 36 according to Kabat numbering, optionally a Tyrosine to Histidine substitution; or
    • (ii) an Alanine at position 46, optionally a substitution at position 46 according to Kabat numbering, optionally a Arginine to Alanine substitution;
    • wherein the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule comprises a light chain comprising a framework region, optionally framework region 3 (FR3), comprising a Phenyalanine at position 87, optionally a substitution at position 87 according to Kabat numbering, optionally a Tyrosine to Phenyalanine substitution, wherein the substitution is relative to a human germline light chain framework region sequence.

In another aspect, provided herein is an antibody molecule which binds, optionally specifically binds, to a T cell receptor beta variable chain (TCRβV) region, wherein the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (a) a light chain variable region (VL) comprising:
    • (i) one, two or all of, optionally three, a light chain complementarity determining region 1 (LC CDR1), a light chain complementarity determining region 2 (LC CDR2), and a light chain complementarity determining region 3 (LC CDR3) of a humanized B-H light chain (LC) of Table 2; and
    • (ii) a framework region (FR) having at least 95% identity with one, two, three or all, optionally four, of a framework region 1 (FR1), a framework region 2 (FR2), a framework region 3 (FR3), and a framework region 4 (FR4) of a humanized B-H LC of Table 2; and/or
    • (b) a heavy chain variable region (VH) comprising:
    • (i) one, two or all of, optionally three, a heavy chain complementarity determining region 1 (HC CDR1), a heavy chain complementarity determining region 2 (HC CDR2) and a heavy chain complementarity determining region 3 (HC CDR3) of a humanized B-H heavy chain (HC) of Table 2; and
    • (ii) a framework region (FR) having at least 95% identity with one, two, three or all, optionally four, of a framework region 1 (FR1), a framework region 2 (FR2), a framework region 3 (FR3), and a framework region 4 (FR4) of a humanized B-H HC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule as provided herein binds to TCRβ V12, optionally TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01, or a variant thereof.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a HC CDR1, a HC CDR2 and a HC CDR3 of Antibody B-H listed in Table 2; or
    • (ii) a LC CDR1, a LC CDR2, and a LC CDR3 of Antibody B-H listed in Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all, optionally three, of a LC CDR1, a LC CDR2 and a LC CDR3 of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO: 11, or an amino acid sequence listed in Table 1.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all, optionally three, of a HC CDR1, a HC CDR2 and a HC CDR3 of a humanized Antibody B-H listed in Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all, optionally three, of a LC CDR1, a LC CDR2 and a LC CDR3 of a humanized Antibody B-H listed in Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • a VH sequence of a humanized Antibody B-H listed in Table 2, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VH of a humanized Antibody B-H listed in Table 2; and/or
    • a VL sequence of a humanized Antibody B-H listed in Table 2, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity to a VL of a humanized Antibody B-H listed in Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with one of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H LC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with any two of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H LC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with any three of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H LC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with all of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H LC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with one of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H HC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with any two of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H HC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with any three of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H HC of Table 2.

In some embodiments, the anti-TCRβV antibody molecule comprises a framework region (FR) having at least 95% identity with all of: a FR1, a FR2, a FR3, and a FR4 of a humanized B-H HC of Table 2.

In some embodiments, binding of the anti-TCRβV antibody molecule to the TCRβV region results in a cytokine profile that differs from a cytokine profile of a T cell engager that binds to a receptor or molecule other than a TCRβV region (“a non-TCRβV-binding T cell engager”).

In some embodiments, the non-TCRβV-binding T cell engager comprises an antibody that binds to a CD3 molecule, optionally CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments, the cytokine profile of the first moiety comprises, one, two, three, four, five, six, seven, or all of the following:

    • (i) increased level, optionally expression level, and/or activity of IL-2;
    • (ii) reduced level, optionally expression level, and/or activity of IL-10;
    • (iii) reduced level, optionally expression level, and/or activity of IL-6;
    • (iv) reduced level, optionally expression level, and/or activity of TNFα;
    • (v) reduced level, optionally expression level, and/or activity of IL-10;
    • (vi) a delay, optionally at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more hours delay, in increased level, optionally expression level, and/or activity of IL-2;
    • (vii) a delay, optionally at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 hours delay, in increased level, optionally expression level, and/or activity of IFNg; or
    • (viii) increased level, optionally expression level, and/or activity of IL-15,
    • optionally wherein (i)-(vii) are relative to the cytokine profile of the non-TCRβV-binding T cell engager.

In some embodiments, binding of the anti-TCRβV antibody molecule to the TCRβV region results in reduced cytokine storm, optionally reduced cytokine release syndrome (CRS), as measured by an assay of Example 3, optionally relative to the cytokine storm induced by the non-TCRβV-binding T cell engager.

In some embodiments, binding of the anti-TCRβV antibody molecule to the TCRβV region results in one, two, three or all of:

    • (ix) reduced T cell proliferation kinetics;
    • (x) cell killing, optionally target cell killing, optionally cancer cell killing, optionally as measured by an assay of Example 4;
    • (xi) increased Natural Killer (NK) cell proliferation, optionally expansion; or
    • (xii) expansion, optionally at least about 1.1-10 fold expansion, optionally at least about 1.1, 1.2, 1.3, 1.4, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, or 10 fold expansion, of a population of T cells having a memory-like phenotype,
    • optionally relative to the non-TCRβV-binding T cell engager.

In some embodiments, the population of T cells having a memory-like phenotype comprises CD45RA+ CCR7− T cells, optionally CD4+ and/or CD8+ T cells.

In some embodiments, binding of the anti-TCRβV antibody molecule to a TCRβV region results in a reduction of at least 2, 5, 10, 20, 50, 100, or 200 fold, or at least 2-200 fold, optionally 5-150, 10-100, 20-50 fold, in the expression level and or activity of IL-10 as measured by an assay of Example 3.

In some embodiments, binding of the anti-TCRβV antibody molecule to a TCRβV region results in a reduction of at least 2, 5, 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 fold, or at least 2-1000 fold, optionally 5-900, 10-800, 20-700, 50-600, 100-500, or 200-400 fold, in the expression level and or activity of IL-6 as measured by an assay of Example 3.

In some embodiments, binding of the anti-TCRβV antibody molecule to a TCRβV region results in a reduction of at least 2, 5, 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, or 2000 fold, or at least 2-2000 fold, optionally 5-1000, 10-900, 20-800, 50-700, 100-600, 200-500, or 300-400 fold, in the expression level and or activity of TNFα as measured by an assay of Example 3.

In some embodiments, binding of the anti-TCRβV antibody molecule to a TCRβV region results in an increase of at least 2, 5, 10, 20, 50, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, or 2000 fold, or at least 2-2000 fold, optionally 5-1000, 10-900, 20-800, 50-700, 100-600, 200-500, or 300-400 fold, in the expression level and or activity of IL-2 as measured by an assay of Example 3.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a single chain Fv (scFv) or a Fab.

In some embodiments, the anti-TCRβV antibody molecule binds to a conformational or a linear epitope on the T cell.

In some embodiments, the anti-TCRβV antibody molecule is a full antibody, optionally an antibody that includes at least one, and preferably two, complete heavy chains, and at least one, and preferably two, complete light chains, or an antigen-binding fragment, optionally a Fab, F(ab′)2, Fv, a single chain Fv fragment, a single domain antibody, a diabody (dAb), a bivalent antibody, or bispecific antibody or fragment thereof, a single domain variant thereof, or a camelid antibody.

In some embodiments, the anti-TCRβV antibody molecule comprises one or more heavy chain constant regions chosen from IgG1, IgG2, IgG3, IgGA1, IgGA2, IgG4, IgJ, IgM, IgD, or IgE, or a fragment thereof, optionally as described in Table 3.

In some embodiments, the anti-TCRβV antibody molecule comprises a heavy chain constant region of an IgM or a fragment thereof, optionally wherein the IgM heavy chain constant region comprises the sequence of SEQ ID NO: 73, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments, the anti-TCRβV antibody molecule comprises a heavy chain constant region of an IgJ or a fragment thereof, optionally wherein the IgJ heavy chain constant region comprises the sequence of SEQ ID NO: 76 or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments, the anti-TCRβV antibody molecule comprises a heavy chain constant region of an IgGA1, or a fragment thereof, optionally wherein the IgGA1 heavy chain constant region comprises the sequence of SEQ ID NO: 74, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments, the anti-TCRβV antibody molecule comprises a heavy chain constant region of an IgGA2, or a fragment thereof, optionally wherein the IgGA2 heavy chain constant region comprises a sequence listed in Table 3, optionally SEQ ID NO: 75, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments, the anti-TCRβV antibody molecule comprises a light chain constant region chosen from the light chain constant regions of kappa or lambda, or a fragment thereof, optionally as described in Table 3.

In some embodiments, the anti-TCRβV antibody molecule comprises a light chain constant region of a kappa chain, or a fragment thereof, optionally wherein the kappa chain constant region comprises the sequence of SEQ ID NO: 39, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • (i) one or more heavy chain constant regions comprising a heavy chain constant region chosen from IgG1, IgG2, IgG3, IgGA1, IgGA2, IgG4, IgJ, IgM, IgD, or IgE, or a fragment thereof, optionally as described in Table 3; and
    • (ii) a light chain constant region comprising a light chain constant region chosen from the light chain constant regions of kappa or lambda, or a fragment thereof, optionally as described in Table 3.

In some embodiments, the anti-TCRβV antibody molecule comprises:

    • (i) a heavy chain constant region comprising:
    • (a) an IgM heavy chain constant region or a fragment thereof, comprising the sequence of SEQ ID NO: 73, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto;
    • (b) an IgGA1 heavy chain constant region or a fragment thereof, comprising the sequence of SEQ ID NO: 74, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto; or
    • (c) an IgGA2 heavy chain constant region or a fragment thereof, comprising the sequence of SEQ ID NO: 75, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto; and
    • (ii) a light chain constant region comprising a kappa chain constant region comprising the sequence of SEQ ID NO: 39, or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto, optionally wherein, the anti-TCRβV antibody molecule further comprises an IgJ heavy chain constant region or a fragment thereof, wherein the IgJ heavy chain constant region comprises the sequence of SEQ ID NO: 76 or a sequence with at least 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments, the second moiety is a tumor-targeting moiety.

In some embodiments, the second moiety is a cytokine molecule.

In some embodiments, the second moiety is a stromal modifying moiety.

In some embodiments, the second moiety is an anti-TCRβV antibody molecule other than the first moiety.

In some embodiments, the first and/or second moiety binds to and activates an immune cell, optionally an effector cell.

In some embodiments, the first and/or second moiety binds to, but does not activate an immune cell, optionally an effector cell.

In some embodiments, the second moiety is chosen from an NK cell engager, a T cell engager other than an anti-TCRβV antibody molecule, a B cell engager, a dendritic cell engager, or a macrophage cell engager, or a combination thereof.

In some embodiments, the tumor-targeting moiety comprises an antibody molecule, optionally Fab or scFv, a receptor molecule, optionally a receptor, a receptor fragment or functional variant thereof, or a ligand molecule, optionally a ligand, a ligand fragment or functional variant thereof, or a combination thereof, that binds to a cancer antigen.

In some embodiments, the tumor-targeting moiety binds to a cancer antigen present on a cancer, optionally a hematological cancer, a solid tumor, a metastatic cancer, soft tissue tumor, metastatic lesion, or a combination thereof.

In some embodiments, the cancer antigen is a tumor antigen or stromal antigen, or a hematological antigen.

In some embodiments, the cancer antigen is chosen from: BCMA, CD19, CD20, CD22, FcRH5, PDL1, CD47, gangloside 2 (GD2), prostate stem cell antigen (PSCA), prostate specific membrane antigen (PMSA), prostate-specific antigen (PSA), carcinoembryonic antigen (CEA), Ron Kinase, c-Met, Immature laminin receptor, TAG-72, BING-4, Calcium-activated chloride channel 2, Cyclin-B1, 9D7, Ep-CAM, EphA3, Her2/neu, Telomerase, SAP-1, Survivin, NY-ESO-1/LAGE-1, PRAME, SSX-2, Melan-A/MART-1, Gp100/pmel 17, Tyrosinase, TRP-1/-2, MC1R, β-catenin, BRCA1/2, CDK4, CML66, Fibronectin, p53, Ras, TGF-B receptor, AFP, ETA, MAGE, MUC-1, CA-125, BAGE, GAGE, NY-ESO-1, β-catenin, CDK4, CDC27, αactinin-4, TRP1/gp75, TRP2, gp100, Melan-A/MART1, gangliosides, WT1, EphA3, Epidermal growth factor receptor (EGFR), MART-2, MART-1, MUC1, MUC2, MUM1, MUM2, MUM3, NA88-1, NPM, OA1, OGT, RCC, RUI1, RUI2, SAGE, TRG, TRP1, TSTA, Folate receptor alpha, L1-CAM, CAIX, gpA33, GD3, GM2, VEGFR, Integrins (Integrin alphaVbeta3, Integrin alpha5Beta1), Carbohydrates (Le), IGF1R, EPHA3, TRAILR1, TRAILR2, RANKL, (FAP), TGF-beta, hyaluronic acid, collagen, optionally collagen IV, tenascin C, or tenascin W.

In some embodiments, the tumor-targeting moiety is a BCMA targeting moiety or a FcRH5 targeting moiety.

In some embodiments, the cancer is a solid tumor including but not limited to: pancreatic, optionally pancreatic adenocarcinoma, cancer, breast cancer, colorectal cancer, lung cancer, optionally small or non-small cell lung cancer, skin cancer, ovarian cancer, or liver cancer.

In some embodiments, the cancer is a hematological cancer including, but not limited to: a B-cell or T cell malignancy, optionally Hodgkin's lymphoma, Non-Hodgkin's lymphoma, optionally B cell lymphoma, diffuse large B cell lymphoma, follicular lymphoma, chronic lymphocytic leukemia, mantle cell lymphoma, marginal zone B-cell lymphoma, Burkitt lymphoma, lymphoplasmacytic lymphoma, hairy cell leukemia, acute myeloid leukemia (AML), chronic myeloid leukemia, myelodysplastic syndrome, multiple myeloma, and acute lymphocytic leukemia.

In some embodiments, the cytokine molecule is chosen from interleukin-2 (IL-2), interleukin-7 (IL-7), interleukin-12 (IL-12), interleukin-15 (IL-15), interleukin-18 (IL-18), interleukin-21 (IL-21), or interferon gamma, or a fragment, variant or combination thereof.

In some embodiments, the cytokine molecule is a monomer or a dimer.

In some embodiments, the cytokine molecule further comprises a receptor dimerizing domain, optionally an IL15Ralpha dimerizing domain.

In some embodiments, the cytokine molecule, optionally IL-15) and the receptor dimerizing domain, optionally an IL15Ralpha dimerizing domain, are not covalently linked, optionally are non-covalently associated.

In some embodiments, the multispecific molecule as provided herein further comprises an immunoglobulin constant region, optionally Fc region, chosen from the heavy chain constant regions of IgG1, IgG2, IgG3, IgGA1, IgGA2, IgG4, IgJ, IgM, IgD, or IgE, or a fragment thereof, optionally wherein, the heavy chain constant region comprises the heavy chain constant region of human IgG1, IgG2 or IgG4.

In some embodiments, the immunoglobulin constant region, optionally an Fc region, is linked, optionally covalently linked to, one or more of tumor-targeting moiety, the cytokine molecule, or the stromal modifying moiety.

In some embodiments, an interface of a first and second immunoglobulin chain constant regions, optionally Fc region, is altered, optionally mutated, to increase or decrease dimerization, optionally relative to a non-engineered interface.

In some embodiments, the dimerization of the immunoglobulin chain constant region, optionally Fc region, is enhanced by providing an Fc interface of a first and a second Fc region with one or more of: a paired cavity-protuberance (“knob-in-a hole”), an electrostatic interaction, or a strand-exchange, such that a greater ratio of heteromultimer:homomultimer forms, optionally relative to a non-engineered interface.

In some embodiments, the multispecific molecule as provided herein further comprises a linker, optionally a linker described herein, optionally wherein the linker is selected from: a cleavable linker, a non-cleavable linker, a peptide linker, a flexible linker, a rigid linker, a helical linker, or a non-helical linker.

In another aspect, provided herein is an isolated nucleic acid molecule comprising a nucleotide sequence encoding the anti-TCRβV antibody molecule as provided herein, or a nucleotide sequence having at least 75%, 80%, 85%, 90%, 95%, or 99% identity thereto.

In another aspect, provided herein is an isolated nucleic acid molecule comprising a nucleotide sequence encoding the multispecific molecule as provided herein, or a nucleotide sequence having at least 75%, 80%, 85%, 90%, 95%, or 99% identity thereto.

In another aspect, provided herein is a vector, optionally an expression vector, comprising one or more of the nucleic acid molecules as provided herein.

In another aspect, provided herein is a cell, optionally host cell, comprising the nucleic acid molecule as provided herein, or the vector as provided herein.

In another aspect, provided herein is a method of making, optionally producing or manufacturing, the anti-TCRβV antibody molecule as provided herein, or the multispecific molecule as provided herein, comprising culturing the host cell as provided herein, under suitable conditions, optionally conditions suitable expression of the anti-TCRβV antibody molecule or the multispecific molecule.

In another aspect, provided herein is a pharmaceutical composition comprising the anti-TCRβV antibody molecule as provided herein, or the multispecific molecule as provided herein, and a pharmaceutically acceptable carrier, excipient, or stabilizer.

In another aspect, provided herein is a method of modulating, optionally enhancing, an immune response in a subject comprising administering to the subject an effective amount of an antibody molecule which binds, optionally specifically binds, to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”).

In another aspect, provided herein is a method of modulating, optionally enhancing, an immune response in a subject comprising administering to the subject an effective amount of the multispecific molecule as provided herein.

In some embodiments, the method comprises expanding, optionally increasing the number of, an immune cell population in the subject.

In another aspect, provided herein is a method of expanding, optionally increasing the number of, an immune cell population comprising, contacting the immune cell population with an effective amount of an antibody molecule which binds, optionally specifically binds, to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”).

In another aspect, provided herein is a method of expanding, optionally increasing the number of, an immune cell population comprising, contacting the immune cell population with an effective amount of the multispecific molecule as provided herein.

In some embodiments, the expansion occurs in vivo or ex vivo, optionally in vitro.

In some embodiments, the immune cell population comprises a TCRβV expressing cell, optionally a TCRβV+ cell.

In some embodiments, the TCRβV expressing cell is a T cell, optionally a CD8+ T cell, a CD3+ T cell or a CD4+ T cell.

In some embodiments, the immune cell population comprises a T cell, optionally a CD4 T cell, a CD8 T cell, optionally an effector T cell, a T cell having a memory-like phenotype or a memory T cell, optionally a memory effector T cell, optionally TEM cell, optionally TEMRA cell, or a tumor infiltrating lymphocyte (TIL).

In some embodiments, the immune cell population comprises a T cell, a Natural Killer cell, a B cell, or a myeloid cell.

In some embodiments, the immune cell population is obtained from a healthy subject.

In some embodiments, the immune cell population is obtained from a subject, optionally from an apheresis sample from the subject, having a disease, optionally a cancer, optionally as described herein, optionally wherein the immune cell population comprises a tumor infiltrating lymphocyte (TIL).

In some embodiments, the method results in an expansion of at least 1.1-10 fold, optionally at least 1.1, 1.2, 1.3, 1.4, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, or 10 fold expansion.

In some embodiments, the method as provided herein further comprises contacting the population of cells with an agent that promotes, optionally increases, immune cell expansion.

In some embodiments, the method as provided herein further comprises contacting the population of cells with an immune checkpoint inhibitor, optionally a PD-1 inhibitor.

In some embodiments, the method as provided herein further comprises contacting the population of cells with a 4-1BB (CD127) agonist, optionally an anti-4-1BB antibody.

In some embodiments, the method as provided herein further comprises contacting the population of cells with a non-dividing population of cells, optionally feeder cells, optionally irradiated allogenic human PBMCs.

In some embodiments, the population of cells is expanded in an appropriate media, optionally media described herein, that includes one or more cytokines, optionally IL-2, IL-7, IL-15, or a combination thereof.

In some embodiments, the population of cells is expanded for a period of at least about 4 hours, 6 hours, 10 hours, 12 hours, 15 hours, 18 hours, 20 hours, or 22 hours, or for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 1,6 17, 18, 19, 20 or 21 days, or for at least about 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks or 8 weeks.

In some embodiments, expansion of the population of immune cells, is compared to expansion of a similar population of cells with an antibody that binds to: a CD3 molecule, optionally CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments, expansion of the population of immune cells, is compared to expansion of a similar population of cells not contacted with the anti-TCRβV antibody molecule, or multi specific molecule comprising the anti-TCRβV antibody molecule.

In some embodiments, expansion of the population of T cells having a memory-like phenotype, optionally CD45RA+ CCR7− cells, optionally memory effector T cells, optionally TEM cells, optionally TEMRA cells, is compared to expansion of a similar population of cells with an antibody that binds to: a CD3 molecule, optionally CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments, the population of expanded T cells having a memory-like phenotype, optionally effector memory cells, comprises cells which:

    • (i) have a detectable level of CD45RA, optionally express or re-express CD45RA;
    • (ii) have low or no expression of CCR7; and/or
    • (iii) have a detectable level of CD95, optionally express CD95,
    • optionally a population of CD45RA+, CCR7−, CD95+ T cells, optionally wherein the T cells comprise CD3+, CD4+ or CD8+ T cells.

In some embodiments, the method results in expansion of, optionally selective or preferential expansion of, T cells expressing a T cell receptor (TCR) comprising a TCR alpha and/or TCR beta molecule, optionally TCR alpha-beta T cells (αβ T cells).

In some embodiments, the method results in expansion of αβT cells over expansion of T cells expressing a TCR comprising a TCR gamma and/or TCR delta molecule, optionally TCR gamma-delta T cells (γδ T cells).

In another aspect, provided herein is a method of treating a disease, optionally cancer, in a subject comprising administering to the subject an effective amount of an antibody molecule which binds, optionally specifically binds, to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”), thereby treating the cancer.

In another aspect, provided herein is a composition comprising an antibody molecule which binds, optionally specifically binds, to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”), for use in treating a disease, optionally cancer, in a subject.

In another aspect, provided herein is a composition comprising an antibody molecule which binds, optionally specifically binds, to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”), for use in the manufacture of a medicament for treating a disease, optionally cancer, in a subject.

In another aspect, provided herein is a method of treating a disease, optionally cancer, in a subject comprising administering to the subject an effective amount of the multispecific molecule as provided herein, thereby treating the cancer.

In another aspect, provided herein is a composition comprising the multispecific molecule as provided herein, for use in treating a disease, optionally cancer, in a subject.

In another aspect, provided herein is a composition comprising the multispecific molecule as provided herein, for use in the manufacture of a medicament for treating a disease, optionally cancer, in a subject.

In another aspect, provided herein is a method of treating, optionally preventing or reducing, cytokine release syndrome (CRS) and/or neurotoxicity (NT) in a subject, optionally CRS and/or NT associated with a treatment, optionally a previously administered treatment, comprising administering to the subject an effective amount of an antibody molecule which binds, optionally specifically binds, to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”), thereby preventing CRS and/or NT in the subject.

In another aspect, provided herein is a method of treating, optionally preventing or reducing, cytokine release syndrome (CRS) and/or neurotoxicity (NT) in a subject, optionally CRS and/or NT associated with a treatment, optionally a previously administered treatment, comprising administering to the subject an effective amount the multispecific molecule as provided herein, thereby preventing CRS and/or NT in the subject.

In another aspect, provided herein is a method of targeting a therapy, optionally treatment, to a T cell in a subject having a disease, optionally cancer, comprising administering an effective amount of:

    • (i) an antibody molecule which binds, optionally specifically binds, to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”); and
    • (ii) the therapy, optionally a tumor targeting therapy, optionally an antibody that binds to a cancer antigen, optionally as described herein, thereby targeting the therapy to the T cell in the subject.

In another aspect, provided herein is a method of targeting a therapy, optionally treatment, to a T cell in a subject having a disease, optionally cancer, comprising administering an effective amount of:

    • (i) the multispecific molecule as provided herein; and
    • (ii) the therapy, optionally a tumor targeting therapy, optionally an antibody that binds to a cancer antigen, optionally as described herein,
    • thereby targeting the therapy to the T cell in the subject.

In some embodiments, the method results in: reduced cytokine release syndrome (CRS), optionally lesser duration of CRS or no CRS, or a reduced severity of CRS, optionally absence of severe CRS, optionally CRS grade 4 or 5, compared to administration of (ii) alone.

In some embodiments, the anti-TCRβV antibody or the multispecific molecule is administered concurrently with or after the administration of the treatment associated with CRS.

In another aspect, provided herein is a method of treating a subject having a cancer, the method comprising:

    • acquiring a value of the status of a TCRβV subfamily for the subject, wherein said value comprises a measure of the presence of, optionally level or activity of, a TCRβV molecule in a sample from the subject, and
    • administering an effective amount of an antibody molecule which binds, optionally specifically binds, to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”); to the subject,
    • thereby treating the subject.

In another aspect, provided herein is a method of treating a subject having a cancer, the method comprising:

    • acquiring a value of the status of a TCRβV subfamily for the subject, wherein said value comprises
    • a measure of the presence of, optionally level or activity of, a TCRβV molecule in a sample from the subject, and
    • administering an effective amount of the multispecific molecule as provided herein to the subject, thereby treating the subject.

In another aspect, provided herein is a method of treating a subject having a cancer, the method comprising administering an effective amount of an antibody molecule which binds, optionally specifically binds, to a T cell receptor beta variable region (TCRβV) (“anti-TCRβV antibody molecule”) to the subject, wherein the subject has a higher, optionally increased, level or activity of one or more TCRβV subfamilies, optionally as described herein, compared to a reference level or activity of one or more TCRβV subfamilies, optionally in a healthy subject, optionally a subject not having a cancer.

In another aspect, provided herein is a method of treating a subject having a cancer, the method comprising administering an effective amount of the multispecific molecule as provided herein to the subject, wherein the subject has a higher, optionally increased, level or activity of one or more TCRβV subfamilies, optionally as described herein, compared to a reference level or activity of one or more TCRβV subfamilies, optionally in a healthy subject, optionally a subject not having a cancer.

In another aspect, provided herein is a method of expanding a population of immune effector cells from a subject having a cancer, the method comprising:

    • (i) isolating a biological sample comprising a population of immune effector cells from the subject; optionally a peripheral blood sample, biopsy sample, or bone marrow sample;
    • (ii) acquiring a value of the status of one or more TCRβV subfamilies for the subject, optionally in the biological sample from the subject, wherein said value comprises a measure of the presence of, optionally level or activity of, a TCRβV subfamily in a sample from the subject compared to a reference value, optionally a sample from a health subject, wherein a value that is higher, optionally increased, in the subject relative to the reference, optionally healthy subject, is indicative of the presence of cancer in the subject, and
    • (iii) contacting the biological sample comprising a population of immune effector cells with an anti-TCRβV antibody molecule, optionally as described herein.

In some embodiments, the method as provided herein further comprises administering the population of immune effector cells contacted with the anti-TCRβV antibody molecule to the subject.

In another aspect, provided herein is a method of expanding a population of immune effector cells from a subject having a cancer, the method comprising:

    • (i) isolating a biological sample comprising a population of immune effector cells from the subject; optionally a peripheral blood sample, biopsy sample, or bone marrow sample;
    • (ii) acquiring a value of the status of one or more TCRβV subfamilies for the subject, optionally in the biological sample from the subject, wherein said value comprises a measure of the presence of, optionally level or activity of, a TCRβV subfamily in a sample from the subject compared to a reference value, optionally a sample from a health subject, wherein a value that is higher, optionally increased, in the subject relative to the reference, optionally healthy subject, is indicative of the presence of cancer in the subject, and
    • (iii) contacting the biological sample comprising a population of immune effector cells with the multispecific molecule as provided herein.

In some embodiments, the method as provided herein further comprises administering the population of immune effector cells contacted with the multispecific molecule to the subject.

In some embodiments, the method as provided herein comprises measuring T cell function, optionally cytotoxic activity, cytokine secretion, or degranulation, in the population of immune effector cells, optionally compared to a reference population, optionally an otherwise similar population not contacted with the anti-TCRβV antibody molecule or a population of immune effector cells obtained from a healthy subject, optionally a subject that does not have a cancer.

In some embodiments, the biological sample comprising the population of immune effector cells is contacted with an anti-TCRβV antibody molecule or a multispecific molecule that binds to the one or more TCRβV subfamilies, optionally the same TCRβV subfamily, identified as being higher, optionally increased, in the biological sample.

In some embodiments, the biological sample comprising the population of immune effector cells is contacted with an anti-TCRβV antibody molecule or a multispecific molecule that does not bind to the one or more TCRβV subfamilies, optionally a different TCRβV subfamily, identified as being higher, optionally increased, in the biological sample.

In some embodiments, the cancer is a solid tumor including but not limited to: melanoma, pancreatic, optionally pancreatic adenocarcinoma, cancer, breast cancer, colorectal cancer (CRC), lung cancer, optionally small or non-small cell lung cancer, skin cancer, ovarian cancer, or liver cancer.

In some embodiments, the cancer is a hematological cancer including, but not limited to: a B-cell or T cell malignancy, optionally Hodgkin's lymphoma, Non-Hodgkin's lymphoma, optionally B cell lymphoma, diffuse large B cell lymphoma (DLBCL), follicular lymphoma, chronic lymphocytic leukemia (B-CLL), mantle cell lymphoma, marginal zone B-cell lymphoma, Burkitt lymphoma, lymphoplasmacytic lymphoma, or hairy cell leukemia, acute myeloid leukemia (AML), chronic myeloid leukemia, myelodysplastic syndrome, multiple myeloma, and acute lymphocytic leukemia.

In some embodiments, the cancer is B-CLL and the TCRβV molecule comprises:

    • (i) TCRβ V6 subfamily comprising, optionally TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01;
    • (ii) TCRβ V5 subfamily comprising TCRβ V5-6*01, TCRβ V5-4*01, or TCRβ V5-8*01;
    • (iii) TCRβ V3 subfamily comprising TCRβ V3-1*01;
    • (iv) TCRβ V2 subfamily comprising TCRβ V2*01; or
    • (v) TCRβ V19 subfamily comprising TCRβ V19*01, or TCRβ V19*02.

In some embodiments, the cancer is melanoma and the TCRβV molecule comprises the TCRβ V6 subfamily comprising, optionally TCRβ V6-4*01, TCRβ V6-4*02, TCRβ V6-9*01, TCRβ V6-8*01, TCRβ V6-5*01, TCRβ V6-6*02, TCRβ V6-6*01, TCRβ V6-2*01, TCRβ V6-3*01 or TCRβ V6-1*01.

In some embodiments, the cancer is DLBCL and the TCRβV molecule comprises:

    • (i) TCRβ V13 subfamily comprising TCRβ V13*01;
    • (ii) TCRβ V3 subfamily comprising TCRβ V3-1*01; or
    • (iii) TCRβ V23 subfamily.

In some embodiments, the cancer is CRC and the TCRβV molecule comprises:

    • (i) TCRβ V19 subfamily comprising TCRβ V19*01, or TCRβ V19*02
    • (ii) TCRβ V12 subfamily comprising TCRβ V12-4*01, TCRβ V12-3*01, or TCRβ V12-5*01
    • (iii) TCRβ V16 subfamily comprising TCRβ V16*01; or
    • (iv) TCRβ V21 subfamily.

In some embodiments, the tumor comprises an antigen, optionally a tumor antigen, optionally a tumor associated antigen or a neoantigen; and/or the one or more TCRβV subfamilies recognize, optionally bind to, the tumor antigen.

In some embodiments, the sample comprises a blood sample, optionally a peripheral blood sample, a biopsy, optionally a tumor biopsy, or a bone marrow sample.

In some embodiments, the sample comprises a biological sample comprising immune cells, optionally TCRBV expressing cells, optionally TCRBV+ cells, T cells, or NK cells.

In some embodiments, the T cells comprise a CD4 T cell, a CD8 T cell, optionally an effector T cell or a memory T cell, optionally a memory effector T cell, optionally TEM cell, optionally TEMRA cell, or a tumor infiltrating lymphocyte (TIL).

In some embodiments, the method results in an expansion, optionally in vivo or ex vivo expansion, of at least 1.1-1000 fold, optionally 1.1-10, 10-100, 100-200, 200-300, 300-400, 400-500, 500-600, 600-700, 700-800, 800-900, or 900-1000 fold expansion of an immune effector cell population comprising a TCRVB expressing immune effector cell, optionally T cell.

In some embodiments, the population of cells is expanded in an appropriate media, optionally media described herein, that includes one or more cytokines, optionally IL-2, IL-7, IL-15, or a combination thereof.

In some embodiments, the population of cells is expanded for a period of at least about 4 hours, 6 hours, 10 hours, 12 hours, 15 hours, 18 hours, 20 hours, or 22 hours, or for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 1,6 17, 18, 19, 20 or 21 days, or for at least about 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks or 8 weeks.

In some embodiments, expansion of the population of immune cells, is compared to expansion of a similar population of cells with an antibody that binds to: a CD3 molecule, optionally CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments, expansion of the population of immune cells, is compared to expansion of a similar population of cells not contacted with the anti-TCRβV antibody molecule.

In some embodiments, expansion of the population of T cells having a memory-like phenotype, optionally memory effector T cells, optionally TEM cells, optionally TEMRA cells, is compared to expansion of a similar population of cells with an antibody that binds to: a CD3 molecule, optionally CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments, the population of expanded T cells having a memory-like phenotype, optionally effector memory cells, comprises cells which:

    • (i) have a detectable level of CD45RA, optionally express or re-express CD45RA;
    • (ii) have low or no expression of CCR7; and/or
    • (iii) have a detectable level of CD95, optionally express CD95,
    • optionally a population of CD45RA+, CCR7−, CD95+ T cells, optionally wherein the T cells comprise CD3+, CD4+ or CD8+ T cells.

In some embodiments, the method results in expansion of, optionally selective or preferential expansion of, T cells expressing a T cell receptor (TCR) comprising a TCR alpha and/or TCR beta molecule, optionally TCR alpha-beta T cells (αβ T cells).

In some embodiments, the method results in expansion of αβT cells over expansion of T cells expressing a TCR comprising a TCR gamma and/or TCR delta molecule, optionally TCR gamma-delta T cells (γδ T cells).

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a light chain variable region (VL) comprising one, two or all of a LC CDR1, a LC CDR2 and a LC CDR3 of a VL disclosed in Tables 1, 2, 10, 11, 12 or 13, optionally SEQ ID NO: 1314, SEQ ID NO: 2, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 16, SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO:30

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising a heavy chain variable region (VH) comprising one, two or all of a HC CDR1, a HC CDR2 and a HC CDR3 of a VH disclosed in Tables 1, 2, 10, 11, 12 or 13, optionally SEQ ID NO: 1312, SEQ ID NO:1, SEQ ID NO: 9, SEQ ID NO: 15, SEQ ID NO: 23, SEQ ID NO: 24 or SEQ ID NO: 25.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • (i) a VL comprising: a LC CDR1 amino acid sequence of SEQ ID NO: 20 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof, a LC CDR2 amino acid sequence of SEQ ID NO:21 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof, and/or a LC CDR3 amino acid sequence of SEQ ID NO:22 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof; and/or
    • (ii) a VH comprising: a HC CDR1 amino acid sequence of SEQ ID NO: 17 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof, a HC CDR2 amino acid sequence of SEQ ID NO:18 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof, and/or a HC CDR3 amino acid sequence of SEQ ID NO:19 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • a variable heavy chain (VH) of SEQ ID NO: 23, SEQ ID NO: 24 or SEQ ID NO: 25, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity thereto; and/or
    • a variable light chain (VL) of SEQ ID NO: 26, SEQ ID NO: 27, SEQ ID NO: 28, SEQ ID NO: 29 or SEQ ID NO:30, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising

    • (i) a VL comprising: a LC CDR1 amino acid sequence of SEQ ID NO: 6 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof, a LC CDR2 amino acid sequence of SEQ ID NO:7 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof, and/or a LC CDR3 amino acid sequence of SEQ ID NO: 8 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof; and/or
    • (ii) a VH comprising: a HC CDR1 amino acid sequence of SEQ ID NO: 3 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof, a HC CDR2 amino acid sequence of SEQ ID NO:4 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof, and/or a HC CDR3 amino acid sequence of SEQ ID NO:5 or an amino acid sequence with not more than 1, 2, 3 or 4 modifications, optionally substitutions, additions or deletions thereof.

In some embodiments, the anti-TCRβV antibody molecule comprises an antigen binding domain comprising:

    • a variable heavy chain (VH) of SEQ ID NO: 1 or SEQ ID NO: 9 or SEQ ID NO: 1312, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity thereto; and/or
    • a variable light chain (VL) of SEQ ID NO: 2, SEQ ID NO: 10 or SEQ ID NO:11 or SEQ ID NO: 1314, or a sequence having at least about 85%, 90%, 95%, or 99% sequence identity thereto.

In some embodiments, the anti-TCRβV antibody molecule comprises a light chain comprising a framework region, optionally framework region 1 (FR1), comprising one, two or all, optionally three, of:

    • (i) an Aspartic Acid at position 1, optionally a substitution at position 1 according to Kabat numbering, optionally a Alanine to Aspartic Acid substitution; or
    • (ii) an Asparagine at position 2, optionally a substitution at position 2 according to Kabat numbering, optionally a Isoleucine to Asparagine, a Serine to Asparagine, or a Tyrosine to Asparagine substitution; or
    • (iii) a Leucine at position 4, optionally a substitution at position 4 according to Kabat numbering, optionally a Methionine to Leucine substitution,
    • wherein the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, the anti-TCRβV antibody molecule comprises a light chain comprising a framework region, optionally framework region 3 (FR3), comprising one, two or all, optionally three, of:

    • (i) a Glycine at position 66, optionally a substitution at position 66 according to Kabat numbering, optionally a Lysine to Glycine, or a Serine to Glycine substitution; or
    • (ii) an Asparagine at position 69, optionally a substitution at position 69 according to Kabat numbering, optionally a Threonine to Asparagine substitution; or
    • (iii) a Tyrosine at position 71, optionally a substitution at position 71 according to Kabat numbering, optionally a Phenylalanine to Tyrosine, or Alanine to Tyrosine substitution,
    • wherein the substitution is relative to a human germline light chain framework region sequence.

In some embodiments, binding of the anti-TCRβV antibody molecule to the TCRβV region results in a cytokine profile that differs from a cytokine profile of a T cell engager that binds to a receptor or molecule other than a TCRβV region (“a non-TCRβV-binding T cell engager”).

In some embodiments, the non-TCRβV-binding T cell engager comprises an antibody that binds to a CD3 molecule, optionally CD3 epsilon (CD3e) molecule; or a TCR alpha (TCRα) molecule.

In some embodiments, the cytokine profile of the first moiety comprises, one, two, three, four, five, six, seven, or all of the following:

    • (i) increased level, optionally expression level, and/or activity of IL-2;
    • (ii) reduced level, optionally expression level, and/or activity of IL-10;
    • (iii) reduced level, optionally expression level, and/or activity of IL-6;
    • (iv) reduced level, optionally expression level, and/or activity of TNFα;
    • (v) reduced level, optionally expression level, and/or activity of IL-10;
    • (vi) a delay, optionally at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more hours delay, in increased level, optionally expression level, and/or activity of IL-2;
    • (vii) a delay, optionally at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 hours delay, in increased level, optionally expression level, and/or activity of IFNg; or
    • (viii) increased level, optionally expression level, and/or activity of IL-15,
    • optionally wherein (i)-(vii) are relative to the cytokine profile of the non-TCRβV-binding T cell engager.

In some embodiments, binding of the anti-TCRβV antibody molecule to the TCRβV region results in reduced cytokine storm, optionally reduced cytokine release syndrome (CRS), as measured by an assay of Example 3, optionally relative to the cytokine storm induced by the non-TCRβV-binding T cell engager.

In some embodiments, binding of the anti-TCRβV antibody molecule to the TCRβV region results in one, two, three or all of:

    • (ix) reduced T cell proliferation kinetics;
    • (x) cell killing, optionally target cell killing, optionally cancer cell killing, optionally as measured by an assay of Example 4;
    • (xi) increased Natural Killer (NK) cell proliferation, optionally expansion; or
    • (xii) expansion, optionally at least about 1.1-10 fold expansion, optionally at least about 1.1, 1.2, 1.3, 1.4, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, or 10 fold expansion, of a population of T cells having a memory-like phenotype,
    • optionally relative to the non-TCRβV-binding T cell engager.

In some embodiments, the anti-TCRβV antibody molecule binds to an outward facing region, optionally epitope, on a TCRβV protein, optionally as depicted by the circled area in FIG. 24A.

In some embodiments, the outward facing region on the TCRβV protein comprises a structurally conserved region of TCRβV, optionally a region of TCRβV having a similar structure across one or more TCRβV subfamilies.

In some embodiments, the method further comprises administering, optionally sequentially, simultaneously or concurrently, a second agent, optionally therapeutic agent, optionally as described herein.

In some embodiments, the second agent, optionally therapeutic agent, comprises a chemotherapeutic agent, a biologic agent, hormonal therapy, radiation, or surgery.

In some embodiments, the disease is a cancer, optionally a solid tumor or a hematological cancer, or a metastatic lesion.

In some embodiments, the cancer antigen is BCMA or FcRH5.

Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In the case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be limiting.

Other features and advantages of the invention will be apparent from the following detailed description and claims.

INCORPORATION BY REFERENCE

All publications and patents mentioned herein are hereby incorporated by reference in their entirety as if each individual publication or patent was specifically and individually indicated to be incorporated by reference.

EQUIVALENTS

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims

1. A composition comprising a molecule that comprises an antigen binding domain that binds to a T cell receptor beta variable (TCRβV) region, wherein the antigen binding domain comprises:

(A) a heavy chain variable region (VH) comprising: (a) a heavy chain complementarity determining region 3 (HC CDR3) comprising the sequence SYYSYDVLDY (SEQ ID NO: 47); and (i) a heavy chain complementarity determining region 1 (HC CDR1) comprising the sequence TYYIH (SEQ ID NO: 45), and a heavy chain complementarity determining region 2 (HC CDR2) comprising the sequence WFFPGSGNIKYNEKFKG (SEQ ID NO: 46); (ii) a HC CDR1 comprising the sequence GHDFRLTYIH (amino acids 26-35 of SEQ ID NO: 1346), and a HC CDR2 comprising the sequence RVSAGSGNVKYNEKFKG (amino acids 50-66 of SEQ ID NO: 1346); (iii) a HC CDR1 comprising the sequence GHDFRLTYIH (amino acids 26-35 of SEQ ID NO: 1356), and a HC CDR2 comprising the sequence RVYAGSGNTKYNEKFKG (amino acids 50-66 of SEQ ID NO: 1356); or (iv) a HC CDR1 comprising the sequence GHDFKLTYIH (amino acids 26-35 of SEQ ID NO: 1370), and a HC CDR2 comprising the sequence RVSAGSGNVNYAQKFQG (amino acids 50-66 of SEQ ID NO: 1370); or (b) a HC CDR3 comprising the sequence of SLNWDYGLDY (SEQ ID NO: 1104); and (i) a HC CDR1 comprising the sequence SAYMH (SEQ ID NO: 1102), and a HC CDR2 comprising the sequence RIDPATGKTKYAPKFQA (SEQ ID NO: 1103); or (ii) a HC CDR1 comprising the sequence DTYMY (SEQ ID NO: 1120); and a HC CDR2 comprising the sequence RIDPANGNTKYDPKFQD (SEQ ID NO: 1121); and
(B) a light chain variable region (VL) comprising: (a) a light chain complementarity determining region 3 (LC CDR3) comprising the sequence QQFKSYPLT (SEQ ID NO: 53); and (i) a light chain complementarity determining region 1 (LC CDR1) comprising the sequence KASQNVGINVV (SEQ ID NO: 51), and a light chain complementarity determining region 2 (LC CDR2) comprising the sequence SSSHRYS (SEQ ID NO: 52); (ii) a LC CDR1 comprising the sequence KASQNVADRVV (amino acids 24-34 of SEQ ID NO: 1349), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349); (iii) a LC CDR1 comprising the sequence KASQNVEDRVV (amino acids 24-34 of SEQ ID NO: 1357), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349); or (iv) a LC CDR1 comprising the sequence RASQNVDNRLG (amino acids 24-34 of SEQ ID NO: 1365), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349); (b) a LC CDR3 comprising the sequence QQFTSSPFT (SEQ ID NO: 65), a LC CDR1 comprising the sequence RASSSVNYIY (SEQ ID NO: 63), and the LC CDR2 comprising the sequence YTSNLAP (SEQ ID NO: 64); (c) a LC CDR3 comprising the sequence QQSIEDPWT (SEQ ID NO: 1109), a LC CDR1 comprising the sequence RASKSVSILGTHLIH (SEQ ID NO: 1107), and the LC CDR2 comprising the sequence AASNLES (SEQ ID NO: 1108); (d) a LC CDR3 comprising the sequence QQSNEDPYT (SEQ ID NO:1128), a LC CDR1 comprising the sequence RASESVDSYGNSFMH (SEQ ID NO: 1126), and the LC CDR2 comprising the sequence RASNLES (SEQ ID NO:1127);

2. The composition of claim 1, wherein the VH comprises:

(i) the HC CDR3 comprising the sequence SYYSYDVLDY (SEQ ID NO: 47);
(ii) the HC CDR1 comprising the sequence TYYIH (SEQ ID NO: 45); and
(iii) the HC CDR2 comprising the sequence WFFPGSGNIKYNEKFKG (SEQ ID NO: 46).

3. The composition of claim 1, wherein the VH comprises:

(i) the HC CDR3 comprising the sequence SYYSYDVLDY (SEQ ID NO: 47);
(ii) the HC CDR1 comprising the sequence GHDFRLTYIH (amino acids 26-35 of SEQ ID NO: 1346); and
(iii) the HC CDR2 comprising the sequence RVSAGSGNVKYNEKFKG (amino acids 50-66 of SEQ ID NO: 1346).

4. The composition of claim 1, wherein the VH comprises:

(i) the HC CDR3 comprising the sequence SYYSYDVLDY (SEQ ID NO: 47);
(ii) the HC CDR1 comprising the sequence GHDFRLTYIH (amino acids 26-35 of SEQ ID NO: 1356); and
(iii) the HC CDR2 comprising the sequence RVYAGSGNTKYNEKFKG (amino acids 50-66 of SEQ ID NO: 1356).

5. The composition of claim 1, wherein the VH comprises:

(i) the HC CDR3 comprising the sequence SYYSYDVLDY (SEQ ID NO: 47);
(ii) the HC CDR1 comprising the sequence GHDFKLTYIH (amino acids 26-35 of SEQ ID NO: 1370); and
(iii) the HC CDR2 comprising the sequence RVSAGSGNVNYAQKFQG (amino acids 50-66 of SEQ ID NO: 1370).

6. The composition of claim 1, wherein the VH comprises:

(i) the HC CDR3 comprising the sequence SLNWDYGLDY (SEQ ID NO: 1104);
(ii) the HC CDR1 comprising the sequence SAYMH (SEQ ID NO: 1102); and
(iii) the HC CDR2 comprising the sequence RIDPATGKTKYAPKFQA (SEQ ID NO: 1103).

7. The composition of claim 1, wherein the VH comprises:

(i) the HC CDR3 comprising the sequence SLNWDYGLDY (SEQ ID NO: 1104);
(ii) the HC CDR1 comprising the sequence DTYMY (SEQ ID NO: 1120); and
(iii) the HC CDR2 comprising the sequence RIDPANGNTKYDPKFQD (SEQ ID NO: 1121).

8. The composition of claim 1, wherein the VL comprises:

(A) (i) the LC CDR3 comprising the sequence QQSIEDPWT (SEQ ID NO:1109); (ii) the LC CDR1 comprising the sequence RASKSVSILGTHLIH (SEQ ID NO:1107); and (iii) the LC CDR2 comprising the sequence AASNLES (SEQ ID NO: 1108);
(B) (i) the LC CDR3 comprising the sequence QQSNEDPYT (SEQ ID NO:1128); (ii) the LC CDR1 comprising the sequence RASESVDSYGNSFMH (SEQ ID NO:1126); and (iii) the LC CDR2 comprising the sequence RASNLES (SEQ ID NO:1127);
(C) (i) the LC CDR3 comprising the sequence QQFKSYPLT (SEQ ID NO: 53); (ii) the LC CDR1 comprising the sequence KASQNVGINVV (SEQ ID NO: 51); and (iii) the LC CDR2 comprising the sequence SSSHRYS (SEQ ID NO: 52);
(D) (i) the LC CDR3 comprising the sequence QQFTSSPFT (SEQ ID NO: 65); (ii) the LC CDR1 comprising the sequence RASSSVNYIY (SEQ ID NO: 63); and (iii) the LC CDR2 comprising the sequence YTSNLAP (SEQ ID NO: 64);
(E) (i) the LC CDR3 comprising the sequence QQFKSYPLT (SEQ ID NO: 53); (ii) the LC CDR1 comprising the sequence KASQNVADRVV (amino acids 24-34 of SEQ ID NO: 1349); and (iii) the LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349);
(F) (i) the LC CDR3 comprising the sequence QQFKSYPLT (SEQ ID NO: 53); (ii) the LC CDR1 comprising the sequence KASQNVEDRVV (amino acids 24-34 of SEQ ID NO: 1357); and (iii) the LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349); or
(G) (i) the LC CDR3 comprising the sequence QQFKSYPLT (SEQ ID NO: 53); (ii) the LC CDR1 comprising the sequence RASQNVDNRLG (amino acids 24-34 of SEQ ID NO: 1365); and (iii) the LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349).

9. The composition of claim 2, wherein the VL comprises:

(i) the LC CDR3 comprising the sequence QQFKSYPLT (SEQ ID NO: 53);
(ii) the LC CDR1 comprising the sequence KASQNVGINVV (SEQ ID NO: 51); and
(iii) the LC CDR2 comprising the sequence SSSHRYS (SEQ ID NO: 52).

10. The composition of claim 3, wherein the VL comprises:

(i) the LC CDR3 comprising the sequence QQFKSYPLT (SEQ ID NO: 53);
(ii) the LC CDR1 comprising the sequence KASQNVADRVV (amino acids 24-34 of SEQ ID NO: 1349); and
(iii) the LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349).

11. The composition of claim 4, wherein the VL comprises:

(i) the LC CDR3 comprising the sequence QQFKSYPLT (SEQ ID NO: 53);
(ii) the LC CDR1 comprising the sequence KASQNVEDRVV (amino acids 24-34 of SEQ ID NO: 1357); and
(iii) the LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349).

12. The composition of claim 5, wherein the VL comprises:

(i) the LC CDR3 comprising the sequence QQFKSYPLT (SEQ ID NO: 53);
(ii) the LC CDR1 comprising the sequence RASQNVDNRLG (amino acids 24-34 of SEQ ID NO: 1365); and
(iii) the LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349).

13. The composition of claim 6, wherein the VL comprises:

(i) the LC CDR3 comprising the sequence QQSIEDPWT (SEQ ID NO:1109);
(ii) a LC CDR1 comprising the sequence RASKSVSILGTHLIH (SEQ ID NO:1107); and
(iii) a LC CDR2 comprising the sequence AASNLES (SEQ ID NO: 1108).

14. The composition of claim 1, wherein the antigen binding domain comprises:

(i) a VH comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1100, and a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1101;
(ii) a VH comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1346, and a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1349;
(iii) a VH comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1356, and a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1357;
(iv) a VH comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1370, and a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1365; or
(v) a VH comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1112, SEQ ID NO: 1113, SEQ ID NO: 1114, SEQ ID NO: 1115, or SEQ ID NO: 1116, and a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1111, SEQ ID NO: 1117, SEQ ID NO: 1118, or SEQ ID NO: 1119.

15. The composition of claim 1, wherein the antigen binding domain comprises:

(i) a VH comprising the sequence of SEQ ID NO: 1100, and a VL comprising the sequence of SEQ ID NO: 1101;
(ii) a VH comprising the sequence of SEQ ID NO: 1346, and a VL comprising the sequence of SEQ ID NO: 1349;
(iii) a VH comprising the sequence of SEQ ID NO: 1356, and a VL comprising the sequence of SEQ ID NO: 1357;
(iv) a VH comprising the sequence of SEQ ID NO: 1370, and a VL comprising the sequence of SEQ ID NO: 1365; or
(v) a VH comprising the sequence of SEQ ID NO: 1112, SEQ ID NO: 1113, SEQ ID NO: 1114, SEQ ID NO: 1115, or SEQ ID NO: 1116, and a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1111, SEQ ID NO: 1117, SEQ ID NO: 1118, or SEQ ID NO: 1119.

16. The composition of claim 1, wherein the antigen binding domain is a Fab or a single chain Fv (scFv).

17. The composition of claim 1, wherein the molecule comprises at least two non-contiguous polypeptide chains,

wherein a first polypeptide chain of the at least two non-contiguous polypeptide chains comprises a first Fc region, and a second polypeptide chain of the at least two non-contiguous polypeptide chains comprises a second Fc region; and
wherein the first Fc region and the second Fc region comprise an Fc interface with a knob-in-a hole.

18. The composition of claim 17, wherein the molecule comprises the first polypeptide chain, the second polypeptide chain, and a third polypeptide chain, wherein the first polypeptide chain, the second polypeptide chain, and the third polypeptide chain are non-contiguous, and wherein:

(i) the first polypeptide chain comprises a VH comprising the sequence of SEQ ID NO: 1346, wherein the VH is linked to the first Fc region,
(ii) the second polypeptide chain comprises a cytokine molecule,
wherein the cytokine molecule is linked to the second Fc region, and
wherein the cytokine molecule comprises: (a) IL-2 or functional variant thereof comprising the sequence of SEQ ID NO: 2270, or (b) IL-15 or functional variant thereof comprising the sequence of SEQ ID NO: 2170, wherein the IL-15 or functional variant thereof is covalently linked to an IL15Ralpha dimerizing domain, and
(iii) the third polypeptide chain comprises a VL comprising the sequence of SEQ ID NO: 1349, wherein the VL is linked to a light chain constant region comprising the sequence of SEQ ID NO: 39.

19. The composition of claim 17, wherein the molecule comprises the first polypeptide chain, the second polypeptide chain, and a third polypeptide chain, wherein the first polypeptide chain, the second polypeptide chain, and the third polypeptide chain are non-contiguous, and wherein:

(i) the first polypeptide chain comprises a VH comprising the sequence of SEQ ID NO: 1113, wherein the VH is linked to the first Fc region,
(ii) the second polypeptide chain comprises a cytokine molecule,
wherein the cytokine molecule is linked to the second Fc region, and
wherein the cytokine molecule comprises: (a) IL-2 or functional variant thereof comprising the sequence of SEQ ID NO: 2270, or (b) IL-15 or functional variant thereof comprising the sequence of SEQ ID NO: 2170, wherein the IL-15 or functional variant thereof is covalently linked to an IL15Ralpha dimerizing domain, and
(iii) the third polypeptide chain comprises a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1111, wherein the VL is linked to a light chain constant region comprising the sequence of SEQ ID NO: 39.

20. The composition of claim 17 wherein the molecule comprises the first polypeptide chain, the second polypeptide chain, and a third polypeptide chain, wherein the first polypeptide chain, the second polypeptide chain, and the third polypeptide chain are non-contiguous, and wherein:

(i) the first polypeptide chain comprises a VH comprising the sequence of SEQ ID NO: 1100, wherein the VH is linked to the first Fc region,
(ii) the second polypeptide chain comprises a cytokine molecule,
wherein the cytokine molecule is linked to the second Fc region, and
wherein the cytokine molecule comprises: (a) IL-2 or functional variant thereof comprising the sequence of SEQ ID NO: 2270, or (b) IL-15 or functional variant thereof comprising the sequence of SEQ ID NO: 2170, wherein the IL-15 or functional variant thereof is covalently linked to an IL15Ralpha dimerizing domain, and
(iii) the third polypeptide chain comprises a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1101, wherein the VL is linked to a light chain constant region comprising the sequence of SEQ ID NO: 39.

21. The composition of claim 17, wherein the molecule comprises the first polypeptide chain, the second polypeptide chain, and a third polypeptide chain, wherein the first polypeptide chain, the second polypeptide chain, and the third polypeptide chain are non-contiguous, and wherein:

(i) the first polypeptide chain comprises a VH comprising the sequence of SEQ ID NO: 1356, wherein the VH is linked to the first Fc region,
(ii) the second polypeptide chain comprises a cytokine molecule,
wherein the cytokine molecule is linked to the second Fc region, and
wherein the cytokine molecule comprises: (a) IL-2 or functional variant thereof comprising the sequence of SEQ ID NO: 2270, or (b) IL-15 or functional variant thereof comprising the sequence of SEQ ID NO: 2170, wherein the IL-15 or functional variant thereof is covalently linked to an IL15Ralpha dimerizing domain, and
(iii) the third polypeptide chain comprises a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1357, wherein the VL is linked to a light chain constant region comprising the sequence of SEQ ID NO: 39.

22. The composition of claim 17, wherein the molecule comprises the first polypeptide chain, the second polypeptide chain, and a third polypeptide chain, wherein the first polypeptide chain, the second polypeptide chain, and the third polypeptide chain are non-contiguous, and wherein:

(i) the first polypeptide chain comprises a VH comprising the sequence of SEQ ID NO: 1370, wherein the VH is linked to the first Fc region,
(ii) the second polypeptide chain comprises a cytokine molecule,
wherein the cytokine molecule is linked to the second Fc region,
wherein the cytokine molecule comprises: (a) IL-2 or functional variant thereof comprising the sequence of SEQ ID NO: 2270, or (b) IL-15 or functional variant thereof comprising the sequence of SEQ ID NO: 2170, wherein the IL-15 or functional variant thereof is covalently linked to an IL15Ralpha dimerizing domain, and
(iii) the third polypeptide chain comprises a VL comprising a sequence having at least 80% sequence identity to SEQ ID NO: 1365, wherein the VL is linked to a light chain constant region comprising the sequence of SEQ ID NO: 39.

23. The composition of claim 18, wherein:

(1) the first Fc region and the second Fc region each comprises an Asn297Ala mutation;
(2) (i) the first Fc region and the second Fc region each comprises a sequence having at least 98% sequence identity to SEQ ID NO: 41 or SEQ ID NO: 42, or (ii) (a) the first Fc region comprising the sequence of amino acids 1-328 of SEQ ID NO: 42, with the proviso that Tyr at the position 349, Thr at the position 366, Leu at the position 368, and Tyr at the position 407 according to EU Numbering are replaced with Cys, Ser, Ala, and Val, respectively; and (b) the second polypeptide chain comprises the cytokine molecule linked to the second Fc region via the sequence of SEQ ID NO: 3308, wherein the second Fc region comprises the sequence of amino acids 104-328 of SEQ ID NO: 42, with the proviso that Ser at the position 354 and Thr at the position 366 according to EU Numbering are replaced with Cys and Trp, respectively; or
(3) any combination thereof.

24. The composition of claim 19, wherein:

(1) the first Fc region and the second Fc region each comprises an Asn297Ala mutation;
(2) (i) the first Fc region and the second Fc region each comprises a sequence having at least 98% sequence identity to SEQ ID NO: 41 or SEQ ID NO: 42, or (ii) (a) the first Fc region comprising the sequence of amino acids 1-328 of SEQ ID NO: 42, with the proviso that Tyr at the position 349, Thr at the position 366, Leu at the position 368, and Tyr at the position 407 according to EU Numbering are replaced with Cys, Ser, Ala, and Val, respectively; and (b) the second polypeptide chain comprises the cytokine molecule linked to the second Fc region via the sequence of SEQ ID NO: 3308, wherein the second Fc region comprises the sequence of amino acids 104-328 of SEQ ID NO: 42, with the proviso that Ser at the position 354 and Thr at the position 366 according to EU Numbering are replaced with Cys and Trp, respectively; or
(3) any combination thereof.

25. The composition of claim 20, wherein:

(1) the first Fc region and the second Fc region each comprises an Asn297Ala mutation;
(2) (i) the first Fc region and the second Fc region each comprises a sequence having at least 98% sequence identity to SEQ ID NO: 41 or SEQ ID NO: 42, or (ii) (a) the first Fc region comprising the sequence of amino acids 1-328 of SEQ ID NO: 42, with the proviso that Tyr at the position 349, Thr at the position 366, Leu at the position 368, and Tyr at the position 407 according to EU Numbering are replaced with Cys, Ser, Ala, and Val, respectively; and (b) the second polypeptide chain comprises the cytokine molecule linked to the second Fc region via the sequence of SEQ ID NO: 3308, wherein the second Fc region comprises the sequence of amino acids 104-328 of SEQ ID NO: 42, with the proviso that Ser at the position 354 and Thr at the position 366 according to EU Numbering are replaced with Cys and Trp, respectively; or
(3) any combination thereof.

26. The composition of claim 21, wherein:

(1) the first Fc region and the second Fc region each comprises an Asn297Ala mutation;
(2) (i) the first Fc region and the second Fc region each comprises a sequence having at least 98% sequence identity to SEQ ID NO: 41 or SEQ ID NO: 42, or (ii) (a) the first Fc region comprising the sequence of amino acids 1-328 of SEQ ID NO: 42, with the proviso that Tyr at the position 349, Thr at the position 366, Leu at the position 368, and Tyr at the position 407 according to EU Numbering are replaced with Cys, Ser, Ala, and Val, respectively; and (b) the second polypeptide chain comprises the cytokine molecule linked to the second Fc region via the sequence of SEQ ID NO: 3308, wherein the second Fc region comprises the sequence of amino acids 104-328 of SEQ ID NO: 42, with the proviso that Ser at the position 354 and Thr at the position 366 according to EU Numbering are replaced with Cys and Trp, respectively; or
(3) any combination thereof.

27. The composition of claim 22, wherein:

(1) the first Fc region and the second Fc region each comprises an Asn297Ala mutation;
(2) (i) the first Fc region and the second Fc region each comprises a sequence having at least 98% sequence identity to SEQ ID NO: 41 or SEQ ID NO: 42, or (ii) (a) the first Fc region comprising the sequence of amino acids 1-328 of SEQ ID NO: 42, with the proviso that Tyr at the position 349, Thr at the position 366, Leu at the position 368, and Tyr at the position 407 according to EU Numbering are replaced with Cys, Ser, Ala, and Val, respectively; and (b) the second polypeptide chain comprises the cytokine molecule linked to the second Fc region via the sequence of SEQ ID NO: 3308, wherein the second Fc region comprises the sequence of amino acids 104-328 of SEQ ID NO: 42, with the proviso that Ser at the position 354 and Thr at the position 366 according to EU Numbering are replaced with Cys and Trp, respectively; or
(3) any combination thereof.

28. The composition of claim 17, wherein the second polypeptide chain comprises the antigen binding domain comprising the sequence of SEQ ID NO: 1331, and a cytokine molecule,

wherein the cytokine molecule comprises IL-2 or functional variant thereof comprising the sequence of SEQ ID NO: 2270, and
wherein the antigen binding domain, the cytokine molecule, and the second Fc region are linked.

29. A method of treating cancer in a human subject in need thereof comprising administering to the human subject a therapeutically effective amount of a molecule comprising an anti-gen binding domain that binds to a T cell receptor beta variable (TCRβV) region, wherein the antigen binding domain comprises:

(A) a heavy chain variable region (VH) comprising: (a) a heavy chain complementarity determining region 3 (HC CDR3) comprising the sequence SYYSYDVLDY (SEQ ID NO: 47); and (i) a heavy chain complementarity determining region 1 (HC CDR1) comprising the sequence TYYIH (SEQ ID NO: 45), and a heavy chain complementarity determining region 2 (HC CDR2) comprising the sequence WFFPGSGNIKYNEKFKG (SEQ ID NO: 46); (ii) a HC CDR1 comprising the sequence GHDFRLTYIH (amino acids 26-35 of SEQ ID NO: 1346), and a HC CDR2 comprising the sequence RVSAGSGNVKYNEKFKG (amino acids 50-66 of SEQ ID NO: 1346); (iii) a HC CDR1 comprising the sequence GHDFRLTYIH (amino acids 26-35 of SEQ ID NO: 1356), and a HC CDR2 comprising the sequence RVYAGSGNTKYNEKFKG (amino acids 50-66 of SEQ ID NO: 1356); or (iv) a HC CDR1 comprising the sequence GHDFKLTYIH (amino acids 26-35 of SEQ ID NO: 1370), and a HC CDR2 comprising the sequence RVSAGSGNVNYAQKFQG (amino acids 50-66 of SEQ ID NO: 1370); or (b) a HC CDR3 comprising the sequence of SLNWDYGLDY (SEQ ID NO: 1104); and (i) a HC CDR1 comprising the sequence SAYMH (SEQ ID NO: 1102), and a HC CDR2 comprising the sequence RIDPATGKTKYAPKFQA (SEQ ID NO: 1103); or (ii) a HC CDR1 comprising the sequence DTYMY (SEQ ID NO: 1120); and a HC CDR2 comprising the sequence RIDPANGNTKYDPKFQD (SEQ ID NO: 1121); and
(B) a light chain variable region (VL) comprising: (a) a light chain complementarity determining region 3 (LC CDR3) comprising the sequence QQFKSYPLT (SEQ ID NO: 53); and (i) a light chain complementarity determining region 1 (LC CDR1) comprising the sequence KASQNVGINVV (SEQ ID NO: 51), and a light chain complementarity determining region 2 (LC CDR2) comprising the sequence SSSHRYS (SEQ ID NO: 52); (ii) a LC CDR1 comprising the sequence KASQNVADRVV (amino acids 24-34 of SEQ ID NO: 1349), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349); (iii) a LC CDR1 comprising the sequence KASQNVEDRVV (amino acids 24-34 of SEQ ID NO: 1357), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349); or (iv) a LC CDR1 comprising the sequence RASQNVDNRLG (amino acids 24-34 of SEQ ID NO: 1365), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349); (b) a LC CDR3 comprising the sequence QQFTSSPFT (SEQ ID NO: 65), a LC CDR1 comprising the sequence RASSSVNYIY (SEQ ID NO: 63), and the LC CDR2 comprising the sequence YTSNLAP (SEQ ID NO: 64); (c) a LC CDR3 comprising the sequence QQSIEDPWT (SEQ ID NO: 1109), a LC CDR1 comprising the sequence RASKSVSILGTHLIH (SEQ ID NO: 1107), and the LC CDR2 comprising the sequence AASNLES (SEQ ID NO: 1108); (d) a LC CDR3 comprising the sequence QQSNEDPYT (SEQ ID NO:1128), a LC CDR1 comprising the sequence RASESVDSYGNSFMH (SEQ ID NO: 1126), and the LC CDR2 comprising the sequence RASNLES (SEQ ID NO:1127), thereby treating the cancer in the human subject.

30. A method of expanding an immune cell population comprising contacting the immune cell population with a composition comprising a molecule comprising an antigen binding domain that binds to a T cell receptor beta variable (TCRβV) region,

wherein the antigen binding domain comprises:
(A) a heavy chain variable region (VH) comprising: (a) a heavy chain complementarity determining region 3 (HC CDR3) comprising the sequence SYYSYDVLDY (SEQ ID NO: 47); and (i) a heavy chain complementarity determining region 1 (HC CDR1) comprising the sequence TYYIH (SEQ ID NO: 45), and a heavy chain complementarity determining region 2 (HC CDR2) comprising the sequence WFFPGSGNIKYNEKFKG (SEQ ID NO: 46); (ii) a HC CDR1 comprising the sequence GHDFRLTYIH (amino acids 26-35 of SEQ ID NO: 1346), and a HC CDR2 comprising the sequence RVSAGSGNVKYNEKFKG (amino acids 50-66 of SEQ ID NO: 1346); (iii) a HC CDR1 comprising the sequence GHDFRLTYIH (amino acids 26-35 of SEQ ID NO: 1356), and a HC CDR2 comprising the sequence RVYAGSGNTKYNEKFKG (amino acids 50-66 of SEQ ID NO: 1356); or (iv) a HC CDR1 comprising the sequence GHDFKLTYIH (amino acids 26-35 of SEQ ID NO: 1370), and a HC CDR2 comprising the sequence RVSAGSGNVNYAQKFQG (amino acids 50-66 of SEQ ID NO: 1370); or (b) a HC CDR3 comprising the sequence of SLNWDYGLDY (SEQ ID NO: 1104); and (i) a HC CDR1 comprising the sequence SAYMH (SEQ ID NO: 1102), and a HC CDR2 comprising the sequence RIDPATGKTKYAPKFQA (SEQ ID NO: 1103); or (ii) a HC CDR1 comprising the sequence DTYMY (SEQ ID NO: 1120); and a HC CDR2 comprising the sequence RIDPANGNTKYDPKFQD (SEQ ID NO: 1121); and
(B) a light chain variable region (VL) comprising: (a) a light chain complementarity determining region 3 (LC CDR3) comprising the sequence QQFKSYPLT (SEQ ID NO: 53); and (i) a light chain complementarity determining region 1 (LC CDR1) comprising the sequence KASQNVGINVV (SEQ ID NO: 51), and a light chain complementarity determining region 2 (LC CDR2) comprising the sequence SSSHRYS (SEQ ID NO: 52); (ii) a LC CDR1 comprising the sequence KASQNVADRVV (amino acids 24-34 of SEQ ID NO: 1349), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349); (iii) a LC CDR1 comprising the sequence KASQNVEDRVV (amino acids 24-34 of SEQ ID NO: 1357), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349); or (iv) a LC CDR1 comprising the sequence RASQNVDNRLG (amino acids 24-34 of SEQ ID NO: 1365), and a LC CDR2 comprising the sequence SSSHRYK (amino acids 50-56 of SEQ ID NO: 1349); (b) a LC CDR3 comprising the sequence QQFTSSPFT (SEQ ID NO: 65), a LC CDR1 comprising the sequence RASSSVNYIY (SEQ ID NO: 63), and the LC CDR2 comprising the sequence YTSNLAP (SEQ ID NO: 64); (c) a LC CDR3 comprising the sequence QQSIEDPWT (SEQ ID NO: 1109), a LC CDR1 comprising the sequence RASKSVSILGTHLIH (SEQ ID NO: 1107), and the LC CDR2 comprising the sequence AASNLES (SEQ ID NO: 1108); (d) a LC CDR3 comprising the sequence QQSNEDPYT (SEQ ID NO:1128), a LC CDR1 comprising the sequence RASESVDSYGNSFMH (SEQ ID NO: 1126), and the LC CDR2 comprising the sequence RASNLES (SEQ ID NO:1127),
thereby expanding the immune cell population, and
wherein the expansion occurs in vivo or ex vivo.
Patent History
Publication number: 20230374133
Type: Application
Filed: Jun 26, 2023
Publication Date: Nov 23, 2023
Inventors: Seng-Lai Tan (Sudbury, MA), Brian Edward Vash (Cambridge, MA), Jonathan Hsu (Waltham, MA), Dilini Charmain Gunasekera (Cambridge, MA), Sangeetha Sagar Palakurthi (East Walpole, MA), Andreas Loew (Boston, MA), Madan Katragadda (Acton, MA), Peter Marek (Hudson, MA), Gurkan Guntas (Newton, MA)
Application Number: 18/341,688
Classifications
International Classification: C07K 16/28 (20060101); A61P 37/04 (20060101); A61P 35/00 (20060101);